US10966961B2 - Pyrazole derivatives as bromodomain inhibitors - Google Patents

Pyrazole derivatives as bromodomain inhibitors Download PDF

Info

Publication number
US10966961B2
US10966961B2 US16/489,859 US201816489859A US10966961B2 US 10966961 B2 US10966961 B2 US 10966961B2 US 201816489859 A US201816489859 A US 201816489859A US 10966961 B2 US10966961 B2 US 10966961B2
Authority
US
United States
Prior art keywords
mmol
alkyl
methyl
pyrazole
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/489,859
Other versions
US20190381010A1 (en
Inventor
Stephen John Atkinson
Emmanuel Hubert Demont
Lee Andrew Harrison
Gemma Michele LIWICKI
Simon Christopher Cranko LUCAS
Alexander G. Preston
Jonathan Thomas Seal
Ian David Wall
Robert J. Watson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline Intellectual Property No 2 Ltd
Original Assignee
GlaxoSmithKline Intellectual Property No 2 Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB1703283.0A external-priority patent/GB201703283D0/en
Priority claimed from GBGB1716374.2A external-priority patent/GB201716374D0/en
Application filed by GlaxoSmithKline Intellectual Property No 2 Ltd filed Critical GlaxoSmithKline Intellectual Property No 2 Ltd
Assigned to GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED reassignment GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ATKINSON, STEPHEN JOHN, DEMONT, EMMANUEL HUBERT, HARRISON, LEE ANDREW, LIWICKI, Gemma Michele, LUCAS, Simon Christopher Cranko, PRESTON, ALEXANDER G., SEAL, JONATHAN THOMAS, WALL, IAN DAVID, WATSON, ROBERT J.
Publication of US20190381010A1 publication Critical patent/US20190381010A1/en
Application granted granted Critical
Publication of US10966961B2 publication Critical patent/US10966961B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/433Thidiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention is directed to pyrazole derivatives which are bromodomain inhibitors, pharmaceutical compositions comprising the compounds and the use of the compounds or the compositions in the treatment of various diseases or conditions, for example acute or chronic autoimmune and/or inflammatory conditions, viral infections and cancer.
  • the genomes of eukaryotic organisms are highly organised within the nucleus of the cell.
  • the long strands of duplex DNA are wrapped around an octomer of histone proteins (most usually comprising two copies of histones H2A, H2B, H3 and H4) to form a nucleosome.
  • This basic unit is then further compressed by the aggregation and folding of nucleosomes to form a highly condensed chromatin structure.
  • a range of different states of condensation are possible, and the tightness of this structure varies during the cell cycle, being most compact during the process of cell division.
  • Chromatin structure plays a critical role in regulating gene transcription, which cannot occur efficiently from highly condensed chromatin.
  • the chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the histone tails which extend beyond the core nucleosome structure. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation and SUMOylation. These epigenetic marks are written and erased by specific enzymes, which place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription.
  • Histone acetylation is most usually associated with the activation of gene transcription, as the modification loosens the interaction of the DNA and the histone octomer by changing the electrostatics.
  • specific proteins recognise and bind to acetylated lysine residues within histones to read the epigenetic code.
  • Bromodomains are small ( ⁇ 110 amino acid) distinct domains within proteins that bind to acetylated lysine resides commonly but not exclusively in the context of histones. There is a family of around 50 proteins known to contain bromodomains, and they have a range of functions within the cell.
  • the BET family of bromodomain containing proteins comprises 4 proteins (BRD2, BRD3, BRD4 and BRDT) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction. Numbering from the N-terminal end of each BET protein the tandem bromodomains are typically labelled Binding Domain 1 (BD1) and Binding Domain 2 (BD2) (Chung et al., J Med. Chem., 2011, 54, 3827-3838).
  • BD1 Binding Domain 1
  • BD2 Binding Domain 2
  • PCT patent applications PCT/EP2016/070519, PCT/EP2016/072216 and PCT/EP2016/073532 each describe a series of pyridone derivatives as bromodomain inhibitors.
  • the invention is directed to compounds of formula (I)
  • R 1 is —C 1-3 alkyl or cyclopropyl
  • R 2 is —C 0-3 alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R 5 groups which may be the same or different; or
  • R 2 is —C 0-4 alkyl-heterocyclyl or —(CH 2 ) p O-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R 9 groups which may be the same or different; or
  • R 2 is H, —CH 3 , —C 2-6 alkyl optionally substituted by one, two, three, four or five fluoro, —C 2-6 alkyl OR 6 , —C 2-6 alkylNR 10a R 11a , —(CH 2 ) m SO 2 C 1-3 alkyl, —(CH 2 ) m SO 2 NR 10 R 11 , —(CH 2 ) m C(O)NR 10 R 11 , —(CH 2 ) m CN, —(CH 2 ) m CO 2 R 6 , —(CH 2 ) m NHCO 2 C 1-4 alkyl, —(CH 2 ) m NHC(O)C 1-4 alkyl or —(CH 2 ) n heteroaryl wherein the heteroaryl is optionally substituted by one or two R 7 groups which may be the same or different;
  • R 3 is H, —C 1-4 alkyl, cyclopropyl, —CH 2 F, —C 1-3 alkylOR 6 or —C 1-3 alkylCN;
  • R 4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R 7 groups which may be the same or different;
  • each R 5 is independently halo, —C 0-6 alkyl-R 8 , —O—C 2-6 alkyl-R 8 , —OCH 2 phenyl, —CN or —SO 2 C 1 -3alkyl;
  • R 6 is H or —C 1-4 alkyl
  • each R 7 is independently oxo, halo, —C 1-4 alkyl optionally substituted by one, two or three fluoro, —C 0-3 alkylOR 6 , —OC 2-3 alkylOR 6 , —C 0-3 alkylNR 19 R 11 , —C 0-3 alkyl-CONR 10 R 11 , —CN, —SO 2 —C 1-3 alkyl, —SO 2 NR 19 R 11 or —SO 2 phenyl optionally substituted by —C 1-4 alkyl;
  • R 8 is H, —OR 6 , —NR 10 R 11 or heteroaryl
  • each R 9 is independently halo, —C 1-4 alkyl, cyclopropyl, cyclobutyl, —CH 2 CF 3 , —CH 2 CHF 2 , —CH 2 CH 2 F, —OCH 2 CH 2 OR 6 , —C 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —NHCH 2 CH 2 OR 6 , —NHCO 2 C 1-4 alkyl, oxo, —C(O)R 6 , —C(O)OR 6 or —C(O)NR 19 R 11 ;
  • R 10 and R 11 are each independently selected from H and —C 1-3 alkyl; or R 10 and R 11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C 1-3 alkyl optionally substituted with one, two or three fluorine atoms, —C 2-4 alkylOH, —OH and F;
  • R 10a and R 11a are each independently selected from H and —C 1-3 alkyl
  • n is an integer selected from 2, 3 or 4;
  • n is an integer selected from 0, 1, 2, 3 or 4;
  • p is an integer selected from 2, 3 or 4.
  • bromodomain inhibitors in particular BD2 selective and may be useful in the treatment of various diseases or conditions, for example acute or chronic auto-immune and/or inflammatory conditions, for example rheumatoid arthritis and cancer.
  • the invention is further directed to pharmaceutical compositions comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention is still further directed to methods of treatment of diseases or conditions associated with bromodomains using a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • BD2 refers to Binding Domain 2 of any of the the BET family of proteins BRD2, BRD3, BRD4 or BRDT.
  • Alkyl refers to a saturated hydrocarbon chain having the specified number of carbon atoms.
  • C 1-3 alkyl and C 1-4 alkyl refers to a straight or branched alkyl group having from 1 to 3 or 1 to 4 carbon atoms respectively.
  • C 0-3 alkyl refers to a straight or branched alkyl group having from 0 (i.e. a bond) to 3 carbon atoms. Representative branched alkyl groups have one, two or three branches.
  • alkyl group may form part of a chain, for example, —C 0-3 alkylOR 6 refers to a straight or branched alkyl chain having from 0 (i.e. a bond) to 3 carbon atoms linked to a group R 6 .
  • Alkyl includes, but is not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, t-butyl, pentyl and hexyl.
  • Cycloalkyl refers to a saturated hydrocarbon mono or bicyclic ring or a saturated spiro-linked bicyclic hydrocarbon ring, having 3, 4, 5, 6, 7, 8, 9 or 10 member atoms in the ring.
  • Suitable examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl, spiro[3.3]heptanyl, bicyclo[2.2.1]heptanyl, adamantyl, bicyclo[3.1.0]hexanyl and bicyclo[2.2.2]octanyl.
  • C 3-7 cycloalkyl refers to a saturated hydrocarbon mono or bicyclic ring or a saturated spiro-linked bicyclic hydrocarbon ring, having 3, 4, 5, 6 or 7 member atoms in the ring.
  • Examples of C 3-7 cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl and bicyclo[3.1.0]hexanyl.
  • Halo refers to a halogen radical, for example, fluoro, chloro, bromo, or iodo.
  • Heteroaryl refers to a monocyclic or bicyclic group having 5, 6, 8, 9, 10 or 11 member atoms, including one, two or three heteroatoms independently selected from nitrogen, sulphur and oxygen, wherein at least a portion of the group is aromatic.
  • the point of attachment to the rest of the molecule may be by any suitable carbon or nitrogen atom.
  • heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, benzofuranyl, isobenzofuryl, 2,3-dihydrobenzofuryl, 1,3-benzodioxolyl, dihydrobenzodioxinyl, benzothienyl, benzazepinyl, 2,3,4,5-tetrahydro-1H-benzo[d]azepinyl, indolizinyl, indolyl, indolinyl, isoindolyl, dihydro
  • C 5-6 heteroaryl refers to a monocyclic aromatic group having 5 or 6 member atoms, including 1, 2, 3 or 4 heteroatoms independently selected from nitrogen, sulphur and oxygen. The point of attachment to the rest of the molecule may be by any suitable carbon or nitrogen atom.
  • C 5-6 heteroaryl groups include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl, pyridazinyl, pyrazinyl, pyrimidinyl and triazinyl.
  • Heteroatom refers to a nitrogen, sulfur, or oxygen atom.
  • Heterocyclyl refers to a non-aromatic heterocyclic monocyclic or bicyclic ring system containing 4, 5, 6, 7, 8, 9 or 10 ring member atoms, including one heteroatom and optionally containing a further heteroatom selected from nitrogen, oxygen or sulphur.
  • heterocyclyl groups include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolidinyl, pyrazolinyl, imidazolidinyl, imidazolinyl, oxazolinyl, thiazolinyl, tetrahydrofuranyl, dihydrofuranyl, 1,3-dioxolanyl, piperidinyl, piperazinyl, homopiperazinyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-oxathiolanyl, 1,3-oxathianyl, 1,3-dithianyl, 1,4-oxathiolanyl, 1,4-oxathianyl, 1,4-
  • “4 to 7-membered heterocyclyl” refers to a non-aromatic heterocyclic ring system containing 4, 5, 6 or 7 ring member atoms, including one heteroatom and optionally containing a further heteroatom selected from nitrogen, oxygen or sulphur.
  • Examples of “4 to 7-membered heterocyclyl” groups include, but are not limited to, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl.
  • Member atoms refers to the atom or atoms that form a chain or ring. Where more than one member atom is present in a chain and within a ring, each member atom is covalently bound to an adjacent member atom in the chain or ring. Atoms that make up a substituent group attached to a chain or ring are not member atoms in the chain or ring.
  • “Substituted” in reference to a group indicates that a hydrogen atom attached to a member atom within a group is replaced. It should be understood that the term “substituted” includes the implicit provision that such substitution be in accordance with the permitted valence of the substituted atom and the substituent and that the substitution results in a stable compound (i.e. one that does not spontaneously undergo transformation such as rearrangement, cyclisation, or elimination). In certain embodiments, a single atom may be substituted with more than one substituent as long as such substitution is in accordance with the permitted valence of the atom. Suitable substituents are defined herein for each substituted or optionally substituted group.
  • “Pharmaceutically acceptable” refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable excipient” refers to a pharmaceutically acceptable material, composition or vehicle involved in giving form or consistency to the pharmaceutical composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of formula (I) or a pharmaceutically acceptable salt thereof when administered to a patient are avoided. In addition, each excipient must of course be pharmaceutically acceptable e.g. of sufficiently high purity.
  • the compounds of the invention may exist in solid or liquid form. In the solid state, the compounds of the invention may exist in crystalline or non-crystalline form, or as a mixture thereof.
  • pharmaceutically acceptable solvates may be formed wherein solvent molecules are incorporated into the crystalline lattice during crystallization. Solvates may involve non-aqueous solvents such as ethanol, iso-propyl alcohol, dimethylsulfoxide (DMSO), acetic acid, ethanolamine, and ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice.
  • Hydrates wherein water is the solvent that is incorporated into the crystalline lattice are typically referred to as “hydrates”. Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The invention includes all such solvates.
  • polymorphs may exhibit polymorphism (i.e. the capacity to occur in different crystalline structures). These different crystalline forms are typically known as “polymorphs”.
  • the invention includes such polymorphs. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification.
  • polymorphs may be produced, for example, by changing or adjusting the reaction conditions or reagents, used in making the compound. For example, changes in temperature, pressure, or solvent may result in polymorphs. In addition, one polymorph may spontaneously convert to another polymorph under certain conditions.
  • Polymorphic forms of compounds of formula (I) may be characterized and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, Raman spectra, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid state nuclear magnetic resonance (SSNMR).
  • XRPD X-ray powder diffraction
  • IR infrared
  • Raman spectra Raman spectra
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • SSNMR solid state nuclear magnetic resonance
  • the compounds according to formula (I) may contain one or more asymmetric centres (also referred to as a chiral centres) and may, therefore, exist as individual enantiomers, diastereoisomers, or other stereoisomeric forms, or as mixtures thereof.
  • Chiral centres such as chiral carbon atoms, may also be present in a substituent such as an alkyl group.
  • the stereochemistry of a chiral centre present in formula (I), or in any chemical structure illustrated herein, is not specified, the structure is intended to encompass any stereoisomer and all mixtures thereof.
  • compounds according to formula (I) containing one or more chiral centres may be used as racemic mixtures, enantiomerically-enriched mixtures, or as enantiomerically-pure individual stereoisomers.
  • the present invention encompasses all isomers of the compounds of formula (I) whether as individual isomers isolated such as to be substantially free of the other isomer (i.e. pure) or as mixtures (i.e. racemic mixtures).
  • An individual isomer isolated such as to be substantially free of the other isomer (i.e. pure) may be isolated such that less than 10%, particularly less than about 1%, for example less than about 0.1% of the other isomer is present.
  • Racemic compounds with a single stereocentre are denoted with either no stereochemistry (single bond) or have the annotation (+/ ⁇ ) or rac. Racemic compounds with two or more stereocentres where relative stereochemistry is known are denoted cis or transas drawn in the structure. Resolved single enantiomers with unknown absolute stereochemistry but known relative stereochemistry are referred to with (R* or S*) with the appropriate relative stereochemistry depicted.
  • Individual stereoisomers of a compound according to formula (I) which contain one or more asymmetric centres may be resolved by methods known to those skilled in the art. For example, such resolution may be carried out (1) by formation of diastereoisomeric salts, complexes or other derivatives; (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • stereoisomers may be synthesised by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
  • salts of the compounds of formula (I) are desirably pharmaceutically acceptable.
  • Suitable pharmaceutically acceptable salts can include acid addition salts or base addition salts.
  • a pharmaceutically acceptable salt may be readily prepared by using a desired acid or base as appropriate. The resultant salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • a pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinic, maleic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, aspartic, p-toluenesulphonic, benzenesulphonic, methanesulphonic, ethanesulphonic, naphthalenesulphonic such as 2-naphthalenesulphonic, or hexanoic acid), optionally in a suitable solvent such as an organic solvent, to give the salt which is usually isolated for example by crystallisation and filtration or by evaporation followed by trituration.
  • a suitable inorganic or organic acid such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinic, maleic, acetic,
  • a pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, succinate, maleate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulphonate, benzenesulphonate, methanesulphonate, ethanesulphonate, naphthalenesulphonate (e.g. 2-naphthalenesulphonate) or hexanoate salt.
  • a hydrobromide hydrochloride, sulfate, nitrate, phosphate, succinate, maleate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulphonate, benzene
  • non-pharmaceutically acceptable salts e.g. formates or trifluoroacetates
  • formates or trifluoroacetates may be used, for example in the isolation of the compounds of formula (I), and are included within the scope of this invention.
  • the invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compounds of formula (I).
  • R 1 is —C 1-3 alkyl or cyclopropyl
  • R 2 is —C 0-3 alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R 5 groups which may be the same or different; or
  • R 2 is —C 0-4 alkyl-heterocyclyl or —(CH 2 ) p O-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R 9 groups which may be the same or different; or
  • R 2 is H, —CH 3 , —C 2-6 alkyl optionally substituted by one, two, three, four or five fluoro, —C 2-6 alkyl OR 6 , —C 2-6 alkyl NR 10a R 11a , —(CH 2 ) m SO 2 C 1-3 alkyl, —(CH 2 ) m SO 2 NR 10 R 11 , —(CH 2 ) m C(O)NR 10 R 11 , —(CH 2 ) m CN, —(CH 2 ) m CO 2 R 6 , —(CH 2 ) m NHCO 2 C 1-4 alkyl, —(CH 2 ) m NHC(O)C 1-4 alkyl or —(CH 2 ) n heteroaryl wherein the heteroaryl is optionally substituted by one or two R 7 groups which may be the same or different;
  • R 3 is H, —C 1-4 alkyl, cyclopropyl, —CH 2 F, —C 1-3 alkylOR 6 or —C 1-3 alkylCN;
  • R 4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R 7 groups which may be the same or different;
  • each R 5 is independently halo, —C 0-6 alkyl-R 8 , —O—C 2-6 alkyl-R 8 , —O—CH 2 phenyl, —CN or —SO 2 C 1 -3alkyl;
  • R 6 is H or —C 1-4 alkyl
  • each R 7 is independently oxo, halo, —C 1-4 alkyl optionally substituted by one, two or three fluoro, —C 0-3 alkylOR 6 , —OC 2-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —C 0-3 alkyl-CONR 10 R 11 , —CN, —SO 2 —C 1-3 alkyl, —SO 2 NR 10 R 11 or —SO 2 phenyl optionally substituted by —C 1-4 alkyl
  • R 8 is H, —OR 6 , —NR 10 R 11 or heteroaryl
  • each R 9 is independently halo, —C 1-4 alkyl, cyclopropyl, cyclobutyl, —CH 2 CF 3 , —CH 2 CHF 2 , —CH 2 CH 2 F, —OCH 2 CH 2 OR 6 , —C 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —NHCH 2 CH 2 OR 6 , —NHCO 2 C 1-4 alkyl, oxo, —C(O)R 6 , —C(O)OR 6 or —C(O)NR 10 R 11 ;
  • R 10 and R 11 are each independently selected from H and —C 1-3 alkyl; or R 10 and R 11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C 1-3 alkyl optionally substituted with one, two or three fluorine atoms, —C 2-4 alkylOH, —OH and F;
  • R 10a and R 11a are each independently selected from H and —C 1-3 alkyl
  • n is an integer selected from 2, 3 or 4;
  • n is an integer selected from 0, 1, 2, 3 or 4;
  • p is an integer selected from 2, 3 or 4.
  • R 1 is —C 1-3 alkyl or cyclopropyl
  • R 2 is —C 0-3 alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R 5 groups which may be the same or different; or
  • R 2 is —C 0-4 alkyl-heterocyclyl or —(CH 2 ) p O-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R 9 groups which may be the same or different; or
  • R 2 is H, —CH 3 , —C 2-6 alkyl optionally substituted by up to five fluoro, —C 2-6 alkylOR 6 , —C 2-6 alkylNR 10a R 11a , —(CH 2 ) m SO 2 C 1-3 alkyl, —(CH 2 ) m SO 2 NR 10 R 11 , —(CH 2 ) m C(O)NR 10 R 11 , —(CH 2 ) m CN, —(CH 2 ) m CO 2 R 6 , —(CH 2 ) m NHCO 2 C 1-4 alkyl, —(CH 2 ) m NHC(O)C 1-4 alkyl or —(CH 2 ) n heteroaryl wherein the heteroaryl is optionally substituted by one or two R 7 groups which may be the same or different;
  • R 3 is —H, —C 1-4 alkyl, cyclopropyl, —CH 2 F, —C 1-3 alkylOR 6 or —C 1-3 alkylCN;
  • R 4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R 7 groups which may be the same or different;
  • each R 5 is independently halo, —C 0-6 alkyl-R 8 , —O—C 2-6 alkyl-R 8 , —CN or —SO 2 C 1-3 alkyl;
  • R 6 is H or —C 1-4 alkyl;
  • each R 7 is independently oxo, halo, —C 1-4 alkyl optionally substituted by 1, 2 or 3 fluoro, —C 0-3 alkylOR 6 , —OC 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —C 0-3 alkyl-CONR 10 R 11 , —CN, —SO 2 —C 1-3 alkyl or —SO 2 NR 10 R 11 ;
  • R 8 is H, —OR 6 , —NR 10 R 11 or heteroaryl
  • each R 9 is independently halo, —C 1-4 alkyl, cyclopropyl, cyclobutyl, —CH 2 CF 3 , —CH 2 CHF 2 , —CH 2 CH 2 F, —OCH 2 CH 2 OR 6 , —C 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —NHCH 2 CH 2 OR 6 , —NHCO 2 R 6 , oxo, —C(O)R 6 , —C(O)OR 6 or —C(O)NR 10 R 11 ;
  • R 10 and R 11 are each independently selected from —H and —C 1-3 alkyl; or R 10 and R 11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C 1-3 alkyl optionally substituted with up to 3 fluorine atoms, —C 2-4 alkylOH, —OH and F;
  • R 10a and R 11a are each independently selected from —H and —C 1-3 alkyl
  • n is an integer selected from 2, 3 or 4;
  • n is an integer selected from 0, 1, 2, 3 or 4;
  • p is an integer selected from 2, 3 or 4.
  • R 1 is methyl
  • R 2 is —C 0-3 alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R 5 groups which may be the same or different.
  • R 2 is a —C 0-3 alkyl-C 3-7 cycloalkyl group, wherein the C 3-7 cycloalkyl group is selected from the group consisting of cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl, said groups being optionally substituted with one, two or three R 5 groups which may be the same or different.
  • R 2 is cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl optionally substituted with one, two or three R 5 groups which may be the same or different.
  • R 2 is cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl optionally substituted with one R 5 group selected from methyl, fluoro and —OH.
  • R 2 is a cyclopropyl optionally substituted by one methyl group.
  • R 2 is a cyclohexyl group optionally substituted with a OH group.
  • R 2 is a cyclohexyl group optionally substituted with a methoxy group.
  • R 2 is —C 0-4 alkyl-heterocyclyl or —(CH 2 ) p O-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R 9 groups which may be the same or different.
  • R 2 is —C 0-4 alkyl-heterocyclyl wherein the heterocyclyl is optionally substituted by one or two R 9 groups which may be the same or different.
  • R 2 is —C 0-4 alkyl-heterocyclyl which is -heterocyclyl, —CH 2 CH 2 -heterocyclyl or —CH 2 CH 2 CH 2 -heterocyclyl.
  • R 2 is —C 0-4 alkyl-heterocyclyl wherein the heterocyclyl is selected from oxetanyl, tetrahydrofuranyl, tetrahydro-2/pyranyl, morpholinyl, piperidinyl, piperazinyl, (1 r,5s)-3-oxa bicyclo[3.1.0]hexanyl and (1r,5S)-3-azabicyclo[3.1.0]hexanyl said groups being optionally substituted by one or two R 9 groups which may be the same or different.
  • R 2 is —C 0-4 alkyl-heterocyclyl wherein the heterocyclyl is selected from oxetanyl, tetrahydrofuranyl, tetrahydro-2/pyranyl, morpholinyl, piperidinyl, piperazinyl, (1r,5S)-3-oxabicyclo[3.1.0]hexanyl and (1r,5s)-3-azabicyclo[3.1.0]hexanyl optionally substituted by one or two R 9 groups selected from methyl, —C(O)CH 3 and fluoro.
  • R 2 is —C 0-4 alkyl-heterocyclyl wherein the heterocyclyl optionally substituted by one or two R 9 groups, is selected from:
  • R 2 is H, —CH 3 , C 2-6 alkyl optionally substituted by one, two, three, four or five fluoro, —C 2-6 alkylOR 6 , —C 2-6 alkylNR 19 R 11 , —(CH 2 ) m SO 2 C 1-3 alkyl, —(CH 2 ) m SO 2 NR 19 R 11 , —(CH 2 ) m C(O)NR 19 R 11 , —(CH 2 ) m CN, —(CH 2 ) m CO 2 R 6 , —(CH 2 ) m NHCO 2 C 1-4 alkyl —(CH 2 ) m NHC(O)C 1-4 alkyl or —(CH 2 ) n heteroaryl wherein the heteroaryl is optionally substituted by one or two R 7 groups which may be the same or different.
  • R 2 is selected from methyl, ethyl, propyl, iso-propyl, butyl, —CH 2 CH 2 CH(CH 3 ) 2 , —CH 2 CH(CH 3 ) 2 , —CH 2 CH 2 OR 6 , —CH 2 CH 2 CH 2 OR 6 , —CH 2 CH(CH 3 )OR 6 , —CH 2 CH 2 CH(CH 3 )OR 6 —CH 2 CH 2 CH(CH 3 )NR 10 R 11 , —CH 2 CH 2 CH 2 NR 10 R 11 , —(CH 2 ) m SO 2 CH 3 , —(CH 2 ) m C(O)NHCH 3 , —(CH 2 ) m CN, —(CH 2 ) m CO 2 R 6 , —(CH 2 ) m CF 3 and —(CH 2 ) m NHCO 2 C(CH 3 ) 3 .
  • R 2 is —C 1-6 alkyl selected from methyl, ethyl, propyl, iso-propyl, butyl, —CH 2 CH 2 CH(CH 3 ) 2 and —CH 2 CH(CH 3 ) 2 .
  • R 2 is —C 1-6 alkylOR 6 selected from —CH 2 CH 2 OR 6 , —CH 2 CH 2 CH 2 OR 6 , —CH 2 CH(CH 3 )OR 6 and —CH 2 CH 2 CH(CH 3 )OR 6 .
  • R 2 is —C 1-6 alkylNR 10 R 11 selected from —CH 2 CH 2 CH(CH 3 )NR 10 R 11 and —CH 2 CH 2 CH 2 NR 10 R 11 .
  • R 2 is —(CH 2 ) m SO 2 CH 3 . In another embodiment R 2 is —(CH 2 ) m C(O)NHCH 3 . In another embodiment R 2 is —(CH 2 ) m CN. In another embodiment R 2 is —(CH 2 ) m CO 2 R 6 . In another embodiment R 2 is —(CH 2 ) m CF 3 . In another embodiment R 2 is —(CH 2 ) m NHCO 2 C(CH 3 ) 3 .
  • R 2 is —(CH 2 ) n heteroaryl wherein the heteroaryl is optionally substituted by one or two R 7 groups which may be the same or different.
  • R 2 is —(CH 2 ) n C 5 -6heteroaryl wherein the C 5-6 heteroaryl is selected from furanyl, thienyl, pyrrolyl, triazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridinyl, pyridazinyl, pyrazinyl and pyrimidinyl said groups being optionally substituted by one or two R′ substituents independently selected from halo, C 1-4 alkyl (such as methyl) and —C 0-3 alkylOR 6 .
  • R 2 is —(CH 2 ) n C 5-6 heteroaryl wherein the C 5-6 heteroaryl is pyrazolyl optionally substituted by C 1-4 alkyl or —C 0-3 alkylOR 6 .
  • R 2 is —(CH 2 ) n C 5-6 heteroaryl wherein the optionally substituted C 5-6 heteroaryl group is selected from the group consisting of
  • R 3 is H, methyl, ethyl, —CH 2 F, —CH 2 OH, —CH(OH)CH 3 , —CH 2 OMe or —CH 2 CN. In one embodiment R 3 is H, methyl, ethyl or CH 2 OH.
  • R 4 is phenyl optionally substituted by one, two or three R 7 groups which may be the same or different. In another embodiment R 4 is unsubstituted phenyl. In another embodiment R 4 is phenyl substituted by one or two R 7 groups which may be the same or different selected from halo, —C 1-4 alkyl optionally substituted one, two or three fluoro, —C 0-3 alkylOR 6 , —OC 2 -3alkylOR 6 and CN. In another embodiment R 4 is phenyl substituted by one or two R 7 groups which may be the same or different selected from halo, —C 1-4 alkyl, —C 0-3 alkylOR 6 and CN. In another embodiment R 4 is phenyl substituted by one R 7 groups selected from the group consisting of fluoro, chloro, methyl, cyano and methoxy.
  • R 4 is a heteroaryl group which is pyridyl optionally substituted by one, two or three R 7 groups which may be the same or different.
  • R 4 is a heteroaryl group which is unsubstituted pyridyl.
  • R 4 is a heteroaryl group which is indolyl (e.g 1H-indol-4-yl or 1H-indol-5-yl) optionally substituted by one, two or three R 7 groups which may be the same or different.
  • R 4 is a heteroaryl group which is 1H-indol-4-yl.
  • R 4 is a heteroaryl group which is a pyrrolopyridinyl (e.g. 1H-pyrrolo[2,3,b]pyridinyl or 1H-pyrrolo[2,3,c]pyridinyl)) optionally substituted by one, two or three R 7 groups which may be the same or different.
  • R 4 is a heteroaryl group which is unsubstituted pyrrolopyridinyl.
  • each R 5 is independently halo or —C 0-6 alkyl-R 8 wherein R 8 is H, OR 6 (such as OH) or NR 10 R 11 (such as NH 2 ).
  • R 6 is H, methyl, ethyl or t-butyl.
  • each R 7 is independently oxo, halo, —C 1-4 alkyl optionally substituted by one, two or three fluoro, —C 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , —C 0-3 alkyl-CONR 10 R 11 , —CN, —SO 2 —C 1-3 alkyl or —SO 2 NR 10 R 11 .
  • each R 7 is independently halo, —C 1-4 alkyl optionally substituted by one, two or three fluoro, —C 0-3 alkylOR 6 or CN.
  • each R 7 is independently halo, —C 1-4 alkyl, —C 0-3 alkylOR 6 or CN;
  • R 8 is H, —OH or methoxy.
  • each R 9 is independently halo, C 1-4 alkyl (such as methyl), —C 0-3 alkylOR 6 , —C 0-3 alkylNR 10 R 11 , oxo, or —C(O)R 6 (such as C(O)CH 3 ).
  • m is 2 or 3.
  • n is 0, 1 or 2. In a further embodiment n is 0. In a yet further embodiment n is 2.
  • p is 2 or 3.
  • Compounds of the invention include the compounds of Examples 1 to 261 and salts thereof.
  • Compounds of the invention include the compounds of Examples 1 to 138 and salts thereof.
  • the compound of formula (I) is selected from:
  • the compound of formula (I) is selected from:
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof for use in therapy, in particular in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • the compounds of the invention may possess an improved profile over known BET inhibitors (including properties such as potency, selectivity and/or developability). Certain compounds of the invention may have an advantageous combination of such properties.
  • the compounds of formula (I) and salts thereof are bromodomain inhibitors, and thus are believed to have potential utility in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • Bromodomain inhibitors are believed to be useful in the treatment of a variety of diseases or conditions related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the prevention and treatment of viral infections.
  • Bromodomain inhibitors may be useful in the treatment of a wide variety of acute or chronic autoimmune and/or inflammatory conditions such as rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease (Crohn's disease and ulcerative colitis), asthma, chronic obstructive airways disease, pneumonitis, myocarditis, pericarditis, myositis, eczema, dermatitis (including atopic dermatitis), alopecia, vitiligo, bullous skin diseases, nephritis, vasculitis, hypercholesterolennia, atherosclerosis, Alzheimer's disease, Sjögren's syndrome, sialoadenitis, central retinal vein occlusion, branched retinal vein occlusion, Irvine-
  • the acute or chronic autoimmune and/or inflammatory condition is a disorder of lipid metabolism mediated via the regulation of APO-A1 such as hypercholesterolemia, atherosclerosis or Alzheimer's disease.
  • the acute or chronic autoimmune and/or inflammatory condition is a respiratory disorder such as asthma or chronic obstructive airways disease.
  • the acute or chronic autoimmune and/or inflammatory condition is a systemic inflammatory disorder such as rheumatoid arthritis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis or inflammatory bowel disease (Crohn's disease or Ulcerative colitis).
  • a systemic inflammatory disorder such as rheumatoid arthritis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis or inflammatory bowel disease (Crohn's disease or Ulcerative colitis).
  • the acute or chronic autoimmune and/or inflammatory condition is multiple sclerosis.
  • the acute or chronic autoimmune and/or inflammatory condition is Type I diabetes.
  • the acute or chronic autoimmune and/or inflammatory condition is rheumatoid arthritis.
  • Bromodomain inhibitors may be useful in the treatment of depression.
  • Bromodomain inhibitors may be useful in the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins, such as sepsis, acute sepsis, sepsis syndrome, septic shock, endotoxaemia, systemic inflammatory response syndrome (SIRS), multi-organ dysfunction syndrome, toxic shock syndrome, acute lung injury, ARDS (adult respiratory distress syndrome), acute renal failure, fulminant hepatitis, burns, acute pancreatitis, post-surgical syndromes, sarcoidosis, Herxheimer reactions, encephalitis, myelitis, meningitis, malaria and SIRS associated with viral infections such as influenza, herpes zoster, herpes simplex and coronavirus.
  • the disease or condition which involves an inflammatory response to an infection with bacteria, a virus, fungi, a parasite or their toxins is acute sepsis.
  • Bromodomain inhibitors may be useful in the treatment of conditions associated with ischaemia-reperfusion injury such as myocardial infarction, cerebro-vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
  • ischaemia-reperfusion injury such as myocardial infarction, cerebro-vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
  • Bromodomain inhibitors may be useful in the treatment of cardiovascular diseases such as coronary artery diseases (for example, angina or myocardial infarction), pulmonary arterial hypertension, cerebro-vascular ischaemia (stroke), hypertensive heart disease, rheumatic heart disease, cardiomyopathy, atrial fibrillation, congenital heart disease, endocarditis, aortic aneurysms or peripheral artery disease.
  • cardiovascular diseases such as coronary artery diseases (for example, angina or myocardial infarction), pulmonary arterial hypertension, cerebro-vascular ischaemia (stroke), hypertensive heart disease, rheumatic heart disease, cardiomyopathy, atrial fibrillation, congenital heart disease, endocarditis, aortic aneurysms or peripheral artery disease.
  • Bromodomain inhibitors may be useful in the treatment of fibrotic conditions such as idiopathic pulmonary fibrosis, pulmonary fibrosis, cystic fibrosis, progressive massive fibrosis, renal fibrosis, liver fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), post-operative stricture, keloid scar formation, scleroderma (including morphea and systemic sclerosis), cardiac fibrosis, atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, arthrofibrosis, Dupuytren's contracture, mediastinal, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis and adhesive capsulitis.
  • fibrotic conditions such as idiopathic pulmonary fibrosis, pulmonary fibrosis, cyst
  • Bromodomain inhibitors may be useful in the treatment of viral infections such as herpes simplex infections and reactivations, cold sores, herpes zoster infections and reactivations, chickenpox, shingles, human papilloma virus (HPV), human immunodeficiency virus (HIV), cervical neoplasia, adenovirus infections, including acute respiratory disease, poxvirus infections such as cowpox or smallpox, or African swine fever virus.
  • the viral infection is a HPV infection of skin or cervical epithelia.
  • the viral infection is a latent HIV infection.
  • Bromodomain inhibitors may be useful in the treatment of a wide variety of bone disorders such as osteoporosis, osteopenia, osteoarthritis and ankylosing spondylitis.
  • Bromodomain inhibitors may be useful in the treatment of cancer, including hematological cancers (such as leukaemia, lymphoma and multiple myeloma), epithelial cancers (including lung, breast or colon carcinomas), midline carcinomas, or mesenchymal, hepatic, renal or neurological tumours.
  • hematological cancers such as leukaemia, lymphoma and multiple myeloma
  • epithelial cancers including lung, breast or colon carcinomas
  • midline carcinomas or mesenchymal
  • hepatic renal or neurological tumours.
  • Bromodomain inhibitors may be useful in the treatment of one or more cancers selected from brain cancer (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast cancer, inflammatory breast cancer, colorectal cancer, Wilm's tumor, Ewing's sarcoma, rhabdomyosarcoma, ependymoma, medulloblastoma, colon cancer, head and neck cancer, kidney cancer, lung cancer, liver cancer, melanoma, squamous cell carcinoma, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma cancer, osteosarcoma, giant cell tumor of bone, thyroid cancer, lymphoblastic T-cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymph
  • the cancer is a leukaemia, for example a leukaemia selected from acute monocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia and mixed lineage leukaemia (MLL).
  • the cancer is NUT-midline carcinoma.
  • the cancer is multiple myeloma.
  • the cancer is a lung cancer such as small cell lung cancer (SCLC).
  • SCLC small cell lung cancer
  • the cancer is a neuroblastoma.
  • the cancer is Burkitt's lymphoma.
  • the cancer is cervical cancer.
  • the cancer is esophageal cancer.
  • the cancer is ovarian cancer.
  • the cancer is breast cancer.
  • the cancer is colorectal cancer.
  • the cancer is prostate cancer.
  • the cancer is castration resistant prostate cancer.
  • Bromodomain inhibitors may be useful in the treatment of diseases associated with systemic inflammatory response syndrome, such as sepsis, burns, pancreatitis, major trauma, haemorrhage and ischaemia.
  • the bromodomain inhibitor would be administered at the point of diagnosis to reduce the incidence of: SIRS, the onset of shock, multi-organ dysfunction syndrome, which includes the onset of acute lung injury, ARDS, acute renal, hepatic, cardiac or gastro-intestinal injury and mortality.
  • the bromodomain inhibitor would be administered prior to surgical or other procedures associated with a high risk of sepsis, haemorrhage, extensive tissue damage, SIRS or MODS (multiple organ dysfunction syndrome).
  • the disease or condition for which a bromodomain inhibitor is indicated is sepsis, sepsis syndrome, septic shock and endotoxaemia.
  • the bromodomain inhibitor is indicated for the treatment of acute or chronic pancreatitis.
  • the bromodomain is indicated for the treatment of burns.
  • the present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy.
  • the compound of formula (I) or a pharmaceutically salt thereof can be used in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • the present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of a disease or condition for which a bromodomain inhibitor is indicated.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of acute or chronic auto-immune and/or inflammatory conditions.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of rheumatoid arthritis there is provided.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of conditions associated with ischaemia-reperfusion injury.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cardiovascular diseases.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of fibrotic conditions for use in the treatment of viral infections.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of bone disorders In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cancer. In a further embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases associated with systemic inflammatory response syndrome.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of acute or chronic auto-immune and/or inflammatory conditions.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of rheumatoid arthritis.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of conditions associated with ischaemia-reperfusion injury.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cardiovascular diseases.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of fibrotic conditions.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of viral infections.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cancer.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases associated with systemic inflammatory response syndrome.
  • a method of treating acute or chronic auto-immune and/or inflammatory conditions in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating rheumatoid arthritis in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating conditions associated with ischaemia-reperfusion injury in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating cardiovascular diseases in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating fibrotic conditions in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating viral infections in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating cancer in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating diseases associated with systemic inflammatory response syndrome in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the subject in need thereof is a mammal, particularly a human.
  • the invention further provides for a method for inhibiting a bromodomain containing protein which comprises contacting the bromodomain containing protein with a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the reference to the “treatment” of a particular disease or condition includes the prevention or prophylaxis of such a disease or condition.
  • a compound of formula (I) as well as pharmaceutically acceptable salts thereof may be administered as the raw chemical, it is common to present the active ingredient as a pharmaceutical composition.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof will normally, but not necessarily, be formulated into pharmaceutical compositions prior to administration to a patient. Accordingly, in another aspect there is provided a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more (e.g. two, three, four, five or six) pharmaceutically acceptable excipients.
  • the compounds of formula (I) and pharmaceutically acceptable salts are as described above.
  • the excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical composition including admixing a compound of formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof may be prepared by, for example, admixture at ambient temperature and atmospheric pressure.
  • the pharmaceutical composition can be used in the treatment of any of the conditions described herein.
  • compositions for the treatment of a disease or condition for which a bromodomain inhibitor is indicated comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the compounds of formula (I) are intended for use in pharmaceutical compositions it will be readily understood that they are each preferably provided in substantially pure form, for example, at least 85% pure, especially at least 98% pure (% in a weight for weight basis).
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered more than once a day.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), ocular (including topical, intraocular, subconjunctival, episcleral, sub-Tenon), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • oral including buccal or sublingual
  • rectal inhaled, intranasal
  • topical including buccal, sublingual or transdermal
  • ocular including topical, intraocular, subconjunctival, episcleral, sub-Tenon
  • vaginal or parenteral including subcutaneous, intramuscular, intravenous or intradermal
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • compositions of the invention may be prepared and packaged in bulk form wherein a safe and effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof can be extracted and then given to the patient such as with powders or syrups.
  • the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form wherein each physically discrete unit contains a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical compositions of the invention typically may contain, for example, from 0.25 mg to 1 g, or from 0.5 mg to 500 mg, or from 1 mg to 100 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixers, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols, solutions, and dry powders; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
  • Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen.
  • suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition.
  • certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting of the compound or compounds of formula (I) or pharmaceutically acceptable salts thereof once administered to the subject from one organ, or portion of the body, to another organ, or portion of the body.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance subject compliance.
  • Suitable pharmaceutically-acceptable excipients include the following types of excipients: carriers, diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavouring agents, flavour-masking agents, colouring agents, anti-caking agents, humectants, chelating agents, plasticisers, viscosity increasing agents, antioxidants, preservatives, stabilisers, surfactants, and buffering agents.
  • carriers diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavouring agents, flavour-masking agents, colouring agents, anti-caking agents, humectants, chelating agents, plasticiser
  • Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention.
  • resources that are available to the skilled artisan which describe pharmaceutically-acceptable excipients and may be useful in selecting suitable pharmaceutically-acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives ( Gower Publishing Limited ), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press ).
  • compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences ( Mack Publishing Company ).
  • the pharmaceutical composition is adapted for parenteral administration, particularly intravenous administration.
  • the pharmaceutical composition is adapted for oral administration.
  • the pharmaceutical composition is adapted for topical administration.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions (which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient) and aqueous and non-aqueous sterile suspensions (which may include suspending agents and thickening agents).
  • aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient
  • aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Powders suitable for incorporating into tablets or capsules may be prepared by reducing the compound to a suitable fine size (e.g. by micronisation) and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • Capsules may be made by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrants include starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone
  • a solution retardant such as paraffin
  • a resorption accelerator such as a quaternary salt
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • compositions for oral administration may be designed to provide a modified release profile so as to sustain or otherwise control the release of the therapeutically active agent.
  • dosage unit compositions for oral administration can be microencapsulated.
  • the composition may be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof may be in a particle-size-reduced form e.g. obtained by micronisation.
  • the preferable particle size of the size-reduced (e.g. micronised) compound or salt is defined by a D 50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, emulsions, lotions, powders, solutions, pastes, gels, foams, sprays, aerosols or oils.
  • Such pharmaceutical compositions may include conventional additives which include, but are not limited to, preservatives, solvents to assist drug penetration, co-solvents, emollients, propellants, viscosity modifying agents (gelling agents), surfactants and carriers.
  • a pharmaceutical composition adapted for topical administration which comprises between 0.01-10%, or between 0.01-1% of the compound of formula (I), or a pharmaceutically acceptable salt thereof, by weight of the composition.
  • compositions are preferably applied as a topical ointment, cream, gel, spray or foam.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • Compositions to be administered to the eye will have ophthalmically compatible pH and osmolality.
  • One or more ophthalmically acceptable pH adjusting agents and/or buffering agents can be included in a composition of the invention, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, and sodium lactate; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • Such acids, bases, and buffers can be included in an amount required to maintain pH of the composition in an ophthalmically acceptable range.
  • One or more ophthalmically acceptable salts can be included in the composition in an amount sufficient to bring osmolality of the composition into an ophthalmically acceptable range.
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions.
  • the ocular delivery device may be designed for the controlled release of one or more therapeutic agents with multiple defined release rates and sustained dose kinetics and permeability. Controlled release may be obtained through the design of polymeric matrices incorporating different choices and properties of biodegradable/bioerodable polymers (e.g.
  • EVA ethylene vinyl) acetate
  • HPC hydroxyalkyl cellulose
  • MC methylcellulose
  • HPMC hydroxypropyl methyl cellulose
  • polycaprolactone poly(glycolic) acid
  • poly(lactic) acid, polyanhydride of polymer molecular weights, polymer crystallinity, copolymer ratios, processing conditions, surface finish, geometry, excipient addition and polymeric coatings that will enhance drug diffusion, erosion, dissolution and
  • compositions for ocular delivery also include in situ gellable aqueous composition.
  • a composition comprises a gelling agent in a concentration effective to promote gelling upon contact with the eye or with lacrimal fluid.
  • Suitable gelling agents include but are not limited to thermosetting polymers.
  • the term “in situgellable” as used herein is includes not only liquids of low viscosity that form gels upon contact with the eye or with lacrimal fluid, but also includes more viscous liquids such as semi-fluid and thixotropic gels that exhibit substantially increased viscosity or gel stiffness upon administration to the eye. See, for example, Ludwig (2005) Adv. Drug Deliv. Rev. 3; 57:1595-639, herein incorporated by reference for purposes of its teachings of examples of polymers for use in ocular drug delivery.
  • Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions, gels or dry powders.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is in a particle-size-reduced form e.g. obtained by micronisation.
  • the preferable particle size of the size-reduced (e.g. micronised) compound or salt is defined by a D 50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
  • Aerosol formulations e.g. for inhaled administration, can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non-aqueous solvent.
  • Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler.
  • the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
  • the dosage form comprises an aerosol dispenser
  • it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC).
  • suitable HFC propellants include 1,1,1,2,3,3,3-heptafluoropropane and 1,1,1,2-tetrafluoroethane.
  • the aerosol dosage forms can also take the form of a pump-atomiser.
  • the pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol.
  • the pharmaceutical composition may be a dry powder inhalable composition.
  • a dry powder inhalable composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of formula (I) or a pharmaceutically acceptable salt thereof (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid and/or metal salt of stearic acid such as magnesium or calcium stearate.
  • the dry powder inhalable composition comprises a dry powder blend of lactose e.g. lactose monohydrate and the compound of formula (I) or salt thereof.
  • Such compositions can be administered to the patient using a suitable device such as the DISKUS® device, marketed by GlaxoSmithKline which is for example described in GB 2242134 A.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof may be formulated as a fluid formulation for delivery from a fluid dispenser, for example a fluid dispenser having a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • a fluid dispenser for example a fluid dispenser having a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations.
  • the dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity.
  • a fluid dispenser of the aforementioned type is described and illustrated in International Patent Application Publication No. WO 2005/044354 A1.
  • each dosage unit for oral or parenteral administration preferably contains from 0.01 mg to 3000 mg, more preferably 0.5 mg to 1000 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • Each dosage unit for nasal or inhaled administration preferably contains from 0.001 mg to 50 mg, more preferably 0.01 mg to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • the pharmaceutically acceptable compounds of formula (I) and pharmaceutically acceptable salts thereof can be administered in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day, 0.5 mg to 1000 mg per day or 100 mg to 2500 mg per day, or a nasal or inhaled dose of 0.001 mg to 50 mg per day or 0.01 mg to 5 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same.
  • An effective amount of a salt thereof may be determined as a proportion of the effective amount of the compound of formula (I) per se.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and the use of at least one other therapeutically active agent.
  • the compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order.
  • the amounts of the compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, according to the invention may be used in combination with or include one or more other therapeutic agents, for example selected from antibiotics, anti-virals, glucocorticosteroids, muscarinic antagonists, beta-2 agonists and Vitamin D3 analogues.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof may be used in combination with a further therapeutic agent which is suitable for the treatment of cancer. Examples of such further therapeutic agents are described in Cancer Principles and Practice of Oncology by V. T. Devita and S.
  • therapeutic agents to be used in combination with the compound of formula (I) or a pharmaceutically acceptable salt thereof include, but are not limited to, anti-microtubule agents (such as diterpenoids and vinca alkaloids); platinum coordination complexes; alkylating agents (such as nitrogen mustards, oxazaphosphorines, alkylsulphonates, nitrosoureas, and triazenes); antibiotic agents (such as anthracyclins, actinomycins and bleomycins); topoisomerase II inhibitors (such as epipodophyllotoxins); antimetabolites (such as purine and pyrimidine analogues and anti-folate compounds); topoisomerase I inhibitors (such as camptothecins; hormones and hormonal ana
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof when administered in combination with other therapeutic agents normally administered by the inhaled, intravenous, oral or intranasal route, that the resultant pharmaceutical composition may be administered by the same routes. Alternatively the individual components of the composition may be administered by different routes.
  • the other therapeutic agent(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic agent. It will be clear also that, where appropriate, the therapeutic agents may be used in optically pure form.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable excipient represent a further aspect of the invention.
  • the compounds of the invention may be made by a variety of methods. Any previously defined variable will continue to have the previously defined meaning unless otherwise indicated. Illustrative general synthetic methods are set out in the following schemes, and can be readily adapted to prepare other compounds of the invention. Specific compounds of the invention are prepared in the Examples section.
  • R 1 , R 2 , R 3 and R 4 are as described above and X is either Br, Cl or OH.
  • Step 1 is an amidation of an ester and may be carried out using an appropriate amine of formula R 1 —NH 2 , optionally in the presence of a suitable solvent, such as THF, at a suitable temperature, such as 0° C. to r.t.
  • a suitable solvent such as THF
  • Step 4 is a base-mediated ester hydrolysis and may be carried out using any suitable base, such as lithium hydroxide, optionally in a suitable solvent or mixture of solvents, such as 1,4-dioxane and water, at a suitable temperature, such as room temperature.
  • Step 5 is an amide coupling reaction and may be carried out using an amine reagent, R 2 —NH 2 , in the presence of a suitable tertiary amine, such as triethylamine or DIPEA, in the presence of a suitable amide coupling reactant, such as HATU, in a suitable solvent, such as DCM or DMF, at a suitable temperature, such as room temperature.
  • Step 6 is an acid-mediated ester cleavage and may be carried out using any suitable acid, such as sulfuric acid, optionally in a suitable solvent or mixture of solvents, such as 1,4 dioxane and water, at a suitable temperature, such as heating at reflux.
  • Step 7 is an amide coupling reaction and may be carried out using an amine reagent, R 1 —NH 2 , in the presence of a suitable tertiary amine, such as triethylamine or DIPEA, in the presence of a suitable amide coupling reactant, such as HATU, in a suitable solvent, such as DCM or DMF, at a suitable temperature, such as room temperature.
  • Suitable amine protecting groups include acyl (e.g. acetyl, carbamate (e.g. 2′,2′,2′-trichloroethoxycarbonyl, benzyloxycarbonyl or t-butoxycarbonyl) and arylalkyl (e.g. benzyl), which may be removed by acid mediated cleavage (e.g. using an acid such as hydrochloric acid in 1,4-dioxane or trifluoroacetic acid in dichloromethane) or reductively (e.g.
  • acyl e.g. acetyl, carbamate (e.g. 2′,2′,2′-trichloroethoxycarbonyl, benzyloxycarbonyl or t-butoxycarbonyl) and arylalkyl (e.g. benzyl), which may be removed by acid mediated cleavage (e.g. using an acid such as hydrochloric acid in 1,
  • Suitable amine protecting groups include trifluoroacetyl (—C(O)CF 3 ) which may be removed by base catalysed hydrolysis.
  • the UPLC analysis was conducted on an Acquity UPLC CSH C18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 40° C.
  • the UV detection was a summed signal from wavelength of 210 nm to 350 nm.
  • the UPLC analysis was conducted on an Acquity UPLC CSH C18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 40° C.
  • A 10 mM ammonium hydrogen carbonate in water adjusted to pH10 with ammonia solution
  • the UV detection was a summed signal from wavelength of 210 nm to 350 nm.
  • the UPLC analysis was conducted on an Acquity UPLC CSH C 18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 40° C.
  • the UV detection was a summed signal from wavelength of 210 nm to 350 nm.
  • the UPLC analysis was conducted on an Acquity BEH C18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 35° C.
  • the UPLC analysis was conducted on an Acquity BEH C18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 35° C.
  • the UPLC analysis was conducted on a Xbridge C18 column (150 mm ⁇ 4.6 mm, i.d. 3.5 ⁇ m packing diameter) at 35° C.
  • the UPLC analysis was conducted on an Acquity BEH C 18 column (100 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 35° C.
  • the UPLC analysis was conducted on an Acquity BEH C 18 column (50 mm ⁇ 2.1 mm, i.d. 1.7 ⁇ m packing diameter) at 35° C.
  • MDAP mass-directed autopreparative chromatography
  • the UV detection was an averaged signal from wavelength of 210 nm to 350 nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
  • MDAP Form
  • HPLC analysis was conducted on an Xselect CSH C18 column (150 mm ⁇ 30 mm i.d. 5 ⁇ m packing diameter) at ambient temperature, eluting with 0.1% formic acid in water (Solvent A) and 0.1% formic acid in acetonitrile (Solvent B) using an elution gradient of between 0 and 100% solvent B over 15 or 25 min.
  • the UV detection was an averaged signal from wavelength of 210 nm to 350 nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
  • the UV detection was an averaged signal from wavelength of 210 nm to 350 nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
  • Diisopropyl (E)-diazene-1,2-dicarboxylate (8.24 g, 40.7 mmol) was added dropwise to a suspension of dimethyl 1H-pyrazole-3,5-dicarboxylate (5 g, 27.2 mmol, commercially available from, for example, Fluorochem), (R)-1-phenylethan-1-ol (3.98 g, 32.6 mmol, commercially available from, for example, Sigma Aldrich) and triphenylphosphine polymer bound 3 mmol/g (13.70 g, 40.7 mmol, commercially available from, for example, Sigma Aldrich) in 2-MeTHF (50 mL) at 0° C. under nitrogen.
  • 2-MeTHF 50 mL
  • Methyl (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylate (4.2 g, 14.62 mmol) was dissolved in a mixture of methanol (10 mL) and THF (10 mL), then NaOH (2M aqueous solution, 14.62 mL, 29.2 mmol) was added and the mixture was stirred for 3 h at rt. The solvent was evaporated in vacuo, the residue dissolved in water (30 mL) and washed with ether, then the aqueous layer acidified with 2M HCl to pH 4 and the resulting mixture stirred for 20 min while cooling in an ice bath.
  • reaction mass was diluted with saturated Rochelle's salt solution (120 mL) and stirred for 16 h, then the layers were separated, and the aqueous phase was extracted with DCM (2 ⁇ 100 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to afford the crude product.
  • the crude product was triturated with n-pentane (3 ⁇ 2 mL) then dried under vaccum to afford (1-tosyl-1H-indol-5-yl)methanol (1.70 g, 5.41 mmol, 97% yield) as an off-white solid.
  • Methyl 1-tosyl-1H-pyrrolo[2,3-c]pyridine-4-carboxylate 510 mg, 1.544 mmol was taken up in DCM (15 mL) under nitrogen and cooled to ⁇ 78° C.
  • DIBAL-H (1M in THF, 15.44 mL, 15.44 mmol) was added to the reaction dropwise and it was left to stir at ⁇ 78° C. for 3 h.
  • the reaction was quenched with methanol (15 mL added dropwise) at ⁇ 78° C. and then allowed to warm to rt. Rochelle's salt (saturated in 25 mL of water) was added and the mixture was stirred for 45 min.
  • Methyl 1H-indole-5-carboxylate (4.31 g, 24.60 mmol, commercially available from, for example, Sigma Aldrich) was taken up in DMF (50 mL) and cooled to 0° C. NaH (60% suspension in mineral oil, 1.181 g, 29.5 mmol) was added in small portions, and the reaction was left to stir for 1 h. Tosyl chloride (5.63 g, 29.5 mmol) was added, and the reaction was allowed to warm to rt and stirred overnight. Additional NaH (0.480 g, 60% suspension in mineral oil) was added, and the reaction left to stir at rt for 1 h. The reaction was quenched with water (25 mL).
  • the reaction mixture was extracted with ethyl acetate (2 ⁇ 50 mL) and the combined organics were washed with brine (75 mL), dried with Na 2 SO 4 , filtered and concentrated in vacuo to yield a brown solid.
  • the crude product was applied to a 100 g ULTRA SNAP cartridge in the minimum of DCM and eluted with 0-5% ethyl acetate in DCM. The appropriate fractions were combined and concentrated in vacuo to yield 1-(1-tosyl-1H-indol-5-yl)-ethan-1-one (1.408 g, 4.27 mmol, 68% yield) as a cream solid.
  • Methyl 1-tosyl-1H-indole-5-carboxylate (1.027 g, 3.12 mmol) was taken up in THF (25 mL) under nitrogen, and cooled to ⁇ 78° C. DIBAL-H (1M in THF, 8 mL, 8.00 mmol) was added dropwise over 10 min and the reaction was left to stir at ⁇ 78° C. for 4 h. The reaction was allowed to warm to rt and was left to stir overnight. The reaction was cooled to ⁇ 78° C. and DIBAL-H (1M in THF, 6.5 mL, 6.50 mmol) was added dropwise over 10 min. The reaction was then allowed to warm to rt and was left to stir for 1.5 h.
  • Methyl 1H-indole-4-carboxylate (5 g, 28.5 mmol, commercially available from, for example, Alfa Aesar) was taken up in DMF (50 mL) and cooled in an ice-bath. NaH (60% suspension in mineral oil, 1.370 g, 34.2 mmol) was added portionwise and the reaction left to stir for 30 min. Tosyl chloride (6.53 g, 34.2 mmol) was added and the reaction left to stir and warm up overnight. The reaction was cooled in an ice-bath and carefully quenched with water (200 mL). The mixture was extracted with EtOAc (2 ⁇ 250 mL).
  • Methyl 1-tosyl-1H-indole-4-carboxylate (6.248 g, 18.97 mmol) was taken up in DCM (200 mL) under nitrogen and cooled to ⁇ 78° C. DIBAL-H (1M, 83 mL, 83 mmol) was added slowly over ⁇ 30 min and the reaction left to stir at ⁇ 78° C. for 90 min before being left to stir and warm to rt over the weekend. The reaction was cooled in an ice-bath and slowly quenched with MeOH (2 mL in 0.1 mL aliquots). Saturated Rochelle's salt solution (200 mL) was added and the mixture stirred and allowed to warm to rt for 3 h.
  • Ethyl 1H-pyrrolo[2,3-b]pyridine-4-carboxylate (1.00 g, 5.26 mmol, commercially available from, for example, Alfa Aesar) was taken up in DMF (10 mL) and cooled in an ice bath. NaH (60% suspension in mineral oil, 0.252 g, 6.31 mmol) was added, and the reaction left to stir for 15 min. Tosyl chloride (1.203 g, 6.31 mmol) was added and the reaction was left to warm up to rt, and stirred for 1 h. The reaction was cooled in an ice bath, and quenched with water (5 mL).
  • Ethyl 1-tosyl-1H-pyrrolo[2,3-b]pyridine-4-carboxylate 936 mg, 2.72 mmol was taken up in DCM (25 mL) under nitrogen and cooled to ⁇ 78° C.
  • DIBAL-H (1M in THF, 27.2 mL, 27.2 mmol) was added to the reaction dropwise and the reaction was left to stir at ⁇ 78° C. overnight.
  • the reaction was quenched with MeOH (1 mL added in small portions), and then allowed to warm to rt. Rochelle's salt (saturated in 50 mL of water) was added and the mixture was stirred for 15 min.
  • Methyl 3-(methylcarbamoyl)-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-5-carboxylate (265 mg, 0.426 mmol) was taken up in methanol (2.5 mL) and THF (2.5 mL). 1M LiOH in water (0.852 mL, 0.852 mmol) was added and the mixture stirred at 50° C. for 90 min. The reaction was concentrated in vacuo. The residue was partitioned between ethyl acetate and water (10 mL each).
  • the reaction mixture was partitioned between ethyl acetate and sodium bicarbonate (15 mL each). The organic layer was washed with 2M HCl (15 mL), brine (15 mL), and then dried with Na 2 SO 4 , filtered and concentrated in vacuo to give the crude product as a yellow oil.
  • the crude product was purified by silica gel column chromatography, eluting with 10-60% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to give the desired product (99.7 mg, 0.178 mmol, 46% yield) as a cream solid.
  • Cyclopent-3-en-1-ol (5 g, 59.4 mmol, commercially available from, for example, Astatech) was dissolved in DCM (100 mL) and TBDMS-Cl (8.96 g, 59.4 mmol) and imidazole (4.86 g, 71.3 mmol) were added, then the resulting suspension was stirred at rt over the weekend. The mixture was washed with water (2 ⁇ 100 mL), dried and evaporated in vacuo to give tert-butyl(cyclopent-3-en-1-yloxy)dimethylsilane (12.05 g, 60.7 mmol, 102% yield) as a pale yellow liquid.
  • reaction mixture was concentrated in vacuo to remove DMF and partitioned between ethyl acetate and saturated aq. LiCl solution.
  • the organic layer was separated, washed with brine, dried (hydrophobic frit) and concentrated to give a crude orange oil. This was purified by chromatography on silica gel column, eluting with 0-50% (25% ethanol in ethyl acetate)/ethyl acetate to give the desired product (330 mg, 0.439 mmol, 80% yield) as a pale yellow oil.
  • Inter- mediate 76 N 5 -((1R,5S,6r)-3- Oxabicyclo[3.1.0]hexan-6-yl)-1- ((R)-2-((tert- butyldimethylsilyl)oxy)-1- phenylethyl)-N 3 -methyl-1H- pyrazole-3,5-dicarboxamide 77 N 3 -Methyl-N 5 -((1S,2S)-2- methylcyclopropyl)-1-(1-(1- tosyl-1H-pyrrolo[2,3-b]pyridin- 4-yl)ethyl)-1H-pyrazole-3,5- dicarboxamide 78 N 5 -((1R,5S,6r)-3- Oxabicyclo[3.1.0]hexan-6-yl)- N 3 -methyl-1-((1-tosyl-1H-indol- 5-yl)methyl)-1H-pyrazole-3,5
  • the aqueous layer was acidified with 2N HCl and extracted with ethyl acetate (2 ⁇ 15 mL). The aqueous layer was then extracted with 10% MeOH in DCM (2 ⁇ 15 mL), and the aqueous layer concentrated in vacuo to give the crude residue.
  • the crude product was dissolved in 1:1 DMSO:MeCN (2 mL), filtered and then purified by MDAP (High pH). The appropriate fractions were concentrated in vacuo to yield the title compound (22 mg, 0.051 mmol, 23% yield) as a cream solid.
  • aqueous LiCl (20 mL), Na 2 CO 3 ( ⁇ 1 mL), and ethyl acetate (20 mL); the layers were separated. The aqueous layer was extracted with further ethyl acetate (2 ⁇ 20 mL). The organic layers were combined, back extracted with water (2 ⁇ 10 mL) and filtered through a cartridge fitted with a hydrophobic frit. The filtrate was evaporated in vacuo to give a colourless gum. This was redissolved in dichloromethane and directly applied to the top of a 10 g SNAP silica cartridge and purified by SP4 flash column chromatography (0-60% ethyl acetate/cyclohexane).
  • Cyclopent-3-en-1-ol (10 g, 119 mmol, commercially available from, for example, Fluorochem) was dissolved in a mixture of DMF (100 mL) and THF (50 mL) and cooled in an ice bath under nitrogen, then NaH (60% suspension in mineral oil, 5.71 g, 143 mmol) was added in small portions and the mixture stirred for 30 min, before addition of MeI (9.66 mL, 155 mmol). The resulting suspension was stirred at 0° C. for 2 h, then added to water (500 mL) and extracted with ether (500 mL). The organic layer was washed with water (2 ⁇ 200 mL) and brine (200 mL), dried and evaporated in vacuo to give the desired product as a pale yellow liquid, which was carried through to subsequent reactions without further purification.
  • Methyl 1H-indole-7-carboxylate 1000 mg, 5.71 mmol, commercially available from, for example, Apollo Scientific was taken up in DMF (18 mL) and cooled to 0° C. under nitrogen.
  • Sodium hydride (60% suspension in mineral oil, 287 mg, 7.18 mmol) was added in small portions and the reaction was left to stir for 15 min.
  • Tosyl-Cl 1308 mg, 6.86 mmol was added, and the reaction was allowed to warm to rt and left to stir for 16 h. The reaction mixture was cooled to 0 degrees before the addition of further sodium hydride (60% suspension in mineral oil, 114 mg, 2.85 mmol).
  • the reaction mixture was stirred for 15 min before being warmed to rt.
  • the reaction mixture was stirred for a further 15 min before the addition of a further portion of tosyl-Cl (552 mg, 2.90 mmol).
  • the resulting solution was stirred for 2 h.
  • the reaction mixture was then heated to 60° C. for 1.5 h.
  • the reaction mixture was quenched with water ( ⁇ 6 mL) before being partitioned between sat. aqueous LiCl (100 mL) and ethyl acetate (30 mL). The layers were separated and the aqueous was extracted twice more with ethyl acetate (2 ⁇ 30 mL).
  • Methyl 1H-indazole-4-carboxylate (530 mg, 3.01 mmol, commercially available from, for example, Sigma Aldrich) was taken up in DMF (6.4 mL) and cooled to 0° C. under nitrogen. Sodium hydride (60% suspension in mineral oil, 241 mg, 6.02 mmol) was added in small portions and the reaction was left to stir for 10 min. Tosyl-Cl (775 mg, 4.07 mmol) was added before stirring for 30 min at 0° C. The reaction was allowed to warm to rt and left to stir for 1 h. The reaction mixture was poured onto 250 mL of water before filtering. The precipitate was dried in a vacuum oven overnight to give methyl 1-tosyl-1H-indazole-4-carboxylate (719 mg, 1.959 mmol, 65% yield).
  • the reaction was quenched with methanol when still at ⁇ 78° C. and after allowed to warm to rt.
  • the reaction was diluted with Rochelle's salt solution (10 mL) and stirred for 16 h.
  • the layers were separated, and the aqueous phase was extracted with DCM (3 ⁇ 20 mL).
  • the combined organic layers were dried through a hydrophobic frit, then evaporated in vacuo to give (1-tosyl-1H-indazol-4-yl)methanol (686 mg, 2.042 mmol, 84% yield).
  • 1-(3-Fluoro-2-methylphenyl)ethan-1-one 1000 mg, 6.57 mmol, commercially available from, for example, Alfa Aesar
  • THF 10 mL
  • ethanol 10 mL
  • Sodium borohydride 456 mg, 12.05 mmol
  • the reaction was slowly quenched with 1M HCl until pH 3, before the addition of water.
  • the layers were separated and the aqueous layer extracted twice more with DCM (2 ⁇ 20 mL).
  • the organic layers were back extracted with sodium bicarbonate aq. solution, passed through a hydrophobic frit and concentrated in vacuo to yield a colourless oil as the crude product, 1-(3-fluoro-2-methylphenyl)ethan-1-ol (875 mg, 5.11 mmol, 78% yield).
  • (+/ ⁇ )-Ethyl 3-(benzyloxy)bicyclo[3.1.0]hexane-6-carboxylate (For a preparation, see Intermediate 110, 0.9 g, 3.46 mmol) was dissolved in EtOH (20 mL) and NaOH (5 mL, 10.00 mmol, 2M aqueous) was added, then the mixture was stirred at rt overnight. The solvent was evaporated in vacuo to about half its original volume, then acidified with 2M HCl (aq) to pH 4 and extracted with EtOAc (50 mL).
  • the resulting gum was purified using silica gel column chromatography eluting with a gradient of 0-30% EtOAc:cyclohexane to give tert-butyl ((1R,3r,5S,6s)-3-(benzyloxy)bicyclo[3.1.0]hexan-6-yl)carbamate (0.35 g, 1.154 mmol, 36.2% yield) as a colourless solid and a mixture of diastereomers.
  • (+/ ⁇ )-(trans)-7-methyl-1,4-dioxaspiro[4.5]decan-8-ol (For a preparation, see Intermediate 115, 425 mg, 2.468 mmol) in acetone (7 mL) was added H 2 SO 4 (18 mL, 9.00 mmol, 0.5M) and the solution was stirred for 20 h at rt.
  • the reaction mixture was diluted with water and extracted with EtOAc. The combined organics were filtered through a hydrophobic frit and concentrated in vacuo to give (+/ ⁇ )-(trans)-4-hydroxy-3-methylcyclohexan-1-one (290 mg, 2.265 mmol, 92% yield) as a yellow oil.
  • (+/ ⁇ )-(trans)-4-hydroxy-3-methylcyclohexan-1-one (For a preparation, see Intermediate 116, 1.99 g, 15.53 mmol) was added to a solution of (R)-2-methylpropane-2-sulfinamide (2.304 g, 18.63 mmol) and tetraethoxytitanium (5.43 mL, 25.9 mmol) in THF (30 mL) and the reaction was stirred at 60° C. for 1.5 h and then at 70° C. for 1.5 h. The mixture was cooled to rt and then to ⁇ 78° C.
  • This oil was purified using silica gel column chromatography eluting with a gradient of 10 to 42% of (25% EtOH in AcOEt):cyclohexane to give (S)—N-((1S,3R,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide and (S)—N-((1S,3S,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide (1:1 diasteromeric mixture) (303.5 mg, 1.301 mmol, 8% yield) as a yellow gum which was used directly in the next reaction.
  • (+)-DIP-Cl (1.265 g, 3.94 mmol) was taken up in THF (20 mL) and was cooled to ⁇ 25° C. under N 2 .
  • 1-(3-Fluoro-4-methylphenyl)ethan-1-one (0.5 g, 3.29 mmol, available from commercial suppliers such as Alfa Aesar) in THF (10 mL) was added and the reaction was left to stir overnight as it warmed from ⁇ 25° C. to rt.
  • the reaction was cooled to ⁇ 35° C. and additional (+)-DIP-Cl (1.265 g, 3.94 mmol) in THF (5 mL) was added. The reaction was kept between ⁇ 35 and ⁇ 25° C.
  • DPPA (26.0 mL, 121 mmol) was added to a solution of (+/ ⁇ )-bicyclo[3.1.0]hex-2-ene-6-carboxylic acid, mixture of diastereomers (For a preparation, see Intermediate 121, 10 g, 81 mmol) and Et 3 N (22.46 mL, 161 mmol) in toluene (10 mL) at rt and the mixture was stirred for 30 min, then benzyl alcohol (16.75 mL, 161 mmol) was added and the mixture heated at reflux for 3 h. The solution was diluted with EtOAc and washed with water and sat.
  • This gum was purified using silica gel column chromatography eluting with a gradient of 0-50% EtOAc:cyclohexane to give (+/ ⁇ ) benzyl ((cis)-2-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate (180 mg, 0.728 mmol, 5% yield) as a colourless gum.
  • (+/ ⁇ )-tert-butyl ((trans)-3-methoxycyclopentyl)carbamate (For a preparation, see Intermediate 126, 280 mg, 1.040 mmol) was taken up in a solution of 4M hydrochloric acid in dioxane (2.5 mL, 10.00 mmol) and was stirred for 2 h. The reaction mixture was concentrated in vacuo to give (+/ ⁇ )-(trans)-3-methoxycyclopentan-1-amine hydrochloride (167 mg, 0.991 mmol, 95% yield).
  • Trimethyloxonium tetrafluoroborate (680 mg, 4.60 mmol) was added, and the reaction allowed to warm to rt and stirred rapidly for 20 h.
  • the reaction was diluted with dichloromethane (50 mL) and washed with water (50 mL).
  • the aqueous layer was extracted with dichloromethane (2 ⁇ 25 mL), and the combined organics were washed with aqueous 0.5 M HCl solution (15 mL), saturated sodium bicarbonate solution (20 mL), and brine (30 mL).
  • the organic phase was passed through a hydrophobic frit and concentrated in vacuo to yield a yellow oil.
  • 3-Hydroxyacetophenone (5.0 g, 36.7 mmol, commercially available from, for example, Sigma Aldrich), ethylene carbonate (4.85 g, 55.1 mmol) and potassium carbonate (5.08 g, 36.7 mmol) were mixed in DMF (50 mL) and heated at 120° C. overnight under nitrogen, then the mixture was diluted with water (200 mL) and extracted with EtOAc (2 ⁇ 100 mL). The combined organics were washed with water (200 mL) and brine (200 mL), dried and evaporated in vacuo to give a pale yellow oil.
  • TBDMS-Cl (4.97 g, 33.0 mmol) was added to a mixture of 1-(3-(2-hydroxyethoxy)phenyl)ethan-1-one (For a preparation, see Intermediate 127, 5.4 g, 30.0 mmol) and imidazole (2.45 g, 36.0 mmol) in DCM (50 mL) and the mixture was stirred for 2 h, then allowed to stand over the weekend. The resulting suspension was washed with water (2 ⁇ 100 mL) and the organic layer dried and evaporated in vacuo to give a colourless oil.
  • (+/ ⁇ )-Methyl 1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (For a preparation, see Intermediate 130, 2.6 g, 5.63 mmol) was dissolved in methanol (30 mL) and NaOH (10 mL, 20.00 mmol, 2M in water) was added, then the mixture was stirred at rt for 2 h. The solvent was then evaporated in vacuo.
  • (+/ ⁇ )-1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (For a preparation, see Intermediate 131, 2 g, 4.47 mmol), (1S,2S)-2-methylcyclopropan-1-amine, hydrochloride (0.625 g, 5.81 mmol), HATU (2.209 g, 5.81 mmol) and Et 3 N (1.868 mL, 13.40 mmol) were dissolved in DCM (20 mL) and the mixture was stirred for 2 h, then allowed to stand over the weekend at rt.
  • the crude product remaining after the first MDAP injection was also purified by MDAP (Formic) with fractions containing desired product partitioned between sat. NaHCO 3 solution and DCM. The organic layer was separated and aqueous layer further extracted with DCM (2 ⁇ 20 mL). Combined organic layers were dried (Na 2 SO 4 ) and concentrated in vacuo to give a second batch of N 5 -((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (30 mg, 0.076 mmol, 21% yield) as a white solid.
  • Examples 44-49 were prepared via an array with the following method:—
  • Examples 50-64 were prepared as part of an amide array using the following commercial amines,
  • Boc deprotection of the reaction intermediate corresponding to the coupling of tert-butyl ((1r,4r)-4-(2-aminoethyl)cyclohexyl)carbamate was carried out in DCM (0.5 mL) using 3M HCl in CPME (0.5 mL). A glass microwave vial was used as the reaction vessel. The reaction was left to stir overnight for 22 h at 35° C. Further DCM (0.5 mL) and 3M HCl in CPME (0.5 mL) were added to the reaction mixture and it was left to stir overnight for 18 h at 35° C. The solvent was then removed to dryness under a stream of nitrogen to afford the HCl salt of the free amine.
  • Example 70 1-((1H-Indol-5-yl)methyl)-N 3 -cyclopropyl-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide
  • Example 71 1-((1H-Indol-4-yl)methyl)-N 3 -cyclopropyl-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide
  • Example 72 N 5 -((1R,3S,5S,6r)-3-Hydroxybicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
  • 3-Hydroxybicyclo[3.1.0]hexane-6-carboxylic acid (720 mg, 5.06 mmol) was dissolved in toluene (10 mL) and tert-butanol (10 mL), then Et 3 N (1.412 mL, 10.13 mmol) and diphenyl phosphorazidate (1.637 mL, 7.60 mmol) were added and the mixture was heated at 80° C. overnight. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL), the solvent dried and evaporated to give a pale yellow gum.
  • Example 75 N 5 -((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-((R)-2-hydroxy-1-phenylethyl)-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide
  • N 5 -((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-1-((R)-2-((tert-butyldimethylsilyl)oxy)-1-phenylethyl)-N 5 -methylpyrazole-3,5-dicarboxamide (214 mg, 0.442 mmol) was taken up in 4M hydrochloric acid in dioxane (500 ⁇ L, 2.000 mmol) and left to stir for 1.5 h. The sample was dissolved in 1:1 MeOH:DMSO (2 ⁇ 1 mL) and purified by MDAP (high pH).
  • Example 76 N 5 -((1R,5S,6s)-3-Azabicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
  • Example 77 N 5 -((1R,5S,6s)-3-Acetyl-3-azabicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
  • Example 78 1-((1H-Indol-4-yl)methyl)-N 5 -((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-M-methyl-1H-pyrazole-3,5-dicarboxamide
  • N 5 -((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (99.7 mg, 0.187 mmol) was taken up in THF (4 mL) and methanol (2 mL). Cesium carbonate (304 mg, 0.934 mmol) was added and the reaction was heated to 73° C. and left to stir overnight. The reaction was concentrated in vacuo.
  • Example 79 1-(1H-Indol-4-yl)methyl)-N 5 -((1R,5S,6s)-3-azabicyclo[3.1.0]hexan-6-yl)-N 5 -methyl-1H-pyrazole-3,5-dicarboxamide
  • Example 80 1-(1-(1H-Indol-4-yl)ethyl)-N 5 -((1r,4r)-4-hydroxycyclohexyl)-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide
  • the layers were separated and the aqueous layer was further extracted with ethyl acetate (2 ⁇ 15 mL). The organic layers were combined, dried with a hydrophobic frit and concentrated in vacuo to give 153 mg of crude residue.
  • the crude product was dissolved in 1:1 DMSO:methanol (2 mL) and was purified by MDAP (Formic). The fractions containing desired product were partitioned between sat. NaHCO 3 solution and DCM. The layers were separated and the aqueous layer was further extracted with two further portions of DCM (2 ⁇ 15 mL). The organic layers were combined, dried (hydrophobic frit) and concentrated in vacuo to give the title compound (79 mg, 0.193 mmol, 33% yield).
  • Example 81 1-((S*)-1-(1H-Indol-4-yl)ethyl)-N 3 -((1r,4S)-4-hydroxycyclohexyl)-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide (single enantiomer of unknown configuration at methyl centre)
  • Example 82 1-(1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)ethyl)-N 3 -methyl-N 3 -((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide
  • Example 83 1-((S*)-1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)ethyl)-N 3 -methyl-N 3 -((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide
  • Example 84 1-((1H-Indol-5-yl)methyl)-N 3 -((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N 5 -methyl-1H-pyrazole-3,5-dicarboxamide
  • Example 85 1-(1-(1H-Indol-5-yl)ethyl)-N 3 -((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N 3 -methyl-1H-pyrazole-3,5-dicarboxamide
  • the combined organics were dried with Na 2 SO 4 , filtered and concentrated in vacuo to yield a colourless solid.
  • the crude product was purified by MDAP (Formic).
  • the MDAP did not collect the product, so the waste was concentrated in vacuo and the residue was purified by MDAP (Formic).
  • the MDAP did not collect the product, so the waste was concentrated in vacuo and the residue was purified by MDAP (Formic).

Abstract

The present invention is directed to pyrazole derivatives, pharmaceutical compositions comprising the compounds and the use of the compounds or the compositions in the treatment of various diseases.

Description

This application is a § 371 of International Application No. PCT/EP2018/054733, filed 27 Feb. 2018, which claims the priority of GB 1716374.2, filed 6 Oct. 2017, and GB 1703283.0, filed 1 Mar. 2017.
FIELD OF THE INVENTION
The present invention is directed to pyrazole derivatives which are bromodomain inhibitors, pharmaceutical compositions comprising the compounds and the use of the compounds or the compositions in the treatment of various diseases or conditions, for example acute or chronic autoimmune and/or inflammatory conditions, viral infections and cancer.
BACKGROUND TO THE INVENTION
The genomes of eukaryotic organisms are highly organised within the nucleus of the cell. The long strands of duplex DNA are wrapped around an octomer of histone proteins (most usually comprising two copies of histones H2A, H2B, H3 and H4) to form a nucleosome. This basic unit is then further compressed by the aggregation and folding of nucleosomes to form a highly condensed chromatin structure. A range of different states of condensation are possible, and the tightness of this structure varies during the cell cycle, being most compact during the process of cell division. Chromatin structure plays a critical role in regulating gene transcription, which cannot occur efficiently from highly condensed chromatin. The chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the histone tails which extend beyond the core nucleosome structure. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation and SUMOylation. These epigenetic marks are written and erased by specific enzymes, which place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription.
Histone acetylation is most usually associated with the activation of gene transcription, as the modification loosens the interaction of the DNA and the histone octomer by changing the electrostatics. In addition to this physical change, specific proteins recognise and bind to acetylated lysine residues within histones to read the epigenetic code. Bromodomains are small (˜110 amino acid) distinct domains within proteins that bind to acetylated lysine resides commonly but not exclusively in the context of histones. There is a family of around 50 proteins known to contain bromodomains, and they have a range of functions within the cell.
The BET family of bromodomain containing proteins comprises 4 proteins (BRD2, BRD3, BRD4 and BRDT) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction. Numbering from the N-terminal end of each BET protein the tandem bromodomains are typically labelled Binding Domain 1 (BD1) and Binding Domain 2 (BD2) (Chung et al., J Med. Chem., 2011, 54, 3827-3838).
Chan et al. report that BET bromodomain inhibition suppresses transcriptional responses to cytokine-Jak-STAT signalling in a gene-specific maner in human monocytes, which suggests that BET inhibition reduces inflammation partially through suppression of cytokine activity. (Chan et al., Eur. J. Immunol., 2015, 45: 287-297).
Klein et al. report that the bromodomain protein inhibitor I-BET151 suppresses expression of inflammatory genes and matrix degrading enzymes in rheumatoid arthritis synovial fibroblasts, which suggests a therapeutic potential in the targeting of epigenetic reader proteins in rheumatoid arthritis. (Klein et al., Ann. Rheum. Dis., 2014, 0:1-8).
Park-Min et al. report that I-BET151 that targets bromo and extra-terminal (BET) proteins that ‘read’ chromatin states by binding to acetylated histones, strongly suppresses osteoclastogenesis. (Park-Min et al. Nature Communications, 2014, 5, 5418).
PCT patent applications PCT/EP2016/070519, PCT/EP2016/072216 and PCT/EP2016/073532 each describe a series of pyridone derivatives as bromodomain inhibitors.
SUMMARY OF THE INVENTION
The invention is directed to compounds of formula (I)
Figure US10966961-20210406-C00001

or a salt thereof
wherein:
R1 is —C1-3alkyl or cyclopropyl;
R2 is —C0-3alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R5 groups which may be the same or different; or
R2 is —C0-4alkyl-heterocyclyl or —(CH2)pO-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different; or
R2 is H, —CH3, —C2-6alkyl optionally substituted by one, two, three, four or five fluoro, —C2-6alkyl OR6, —C2-6alkylNR10aR11a, —(CH2)mSO2C1-3alkyl, —(CH2)mSO2NR10R11, —(CH2)mC(O)NR10R11, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mNHCO2C1-4alkyl, —(CH2)mNHC(O)C1-4alkyl or —(CH2)nheteroaryl wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different;
R3 is H, —C1-4alkyl, cyclopropyl, —CH2F, —C1-3alkylOR6 or —C1-3alkylCN;
R4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R7 groups which may be the same or different;
each R5 is independently halo, —C0-6alkyl-R8, —O—C2-6alkyl-R8, —OCH2phenyl, —CN or —SO2C1-3alkyl;
R6 is H or —C1-4alkyl;
each R7 is independently oxo, halo, —C1-4alkyl optionally substituted by one, two or three fluoro, —C0-3alkylOR6, —OC2-3alkylOR6, —C0-3alkylNR19R11, —C0-3alkyl-CONR10R11, —CN, —SO2—C1-3alkyl, —SO2NR19R11 or —SO2phenyl optionally substituted by —C1-4alkyl;
R8 is H, —OR6, —NR10R11 or heteroaryl;
each R9 is independently halo, —C1-4alkyl, cyclopropyl, cyclobutyl, —CH2CF3, —CH2CHF2, —CH2CH2F, —OCH2CH2OR6, —C0-3alkylOR6, —C0-3alkylNR10R11, —NHCH2CH2OR6, —NHCO2C1-4alkyl, oxo, —C(O)R6, —C(O)OR6 or —C(O)NR19R11;
R10 and R11 are each independently selected from H and —C1-3alkyl; or R10 and R11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C1-3alkyl optionally substituted with one, two or three fluorine atoms, —C2-4alkylOH, —OH and F;
R10a and R11a are each independently selected from H and —C1-3alkyl;
m is an integer selected from 2, 3 or 4;
n is an integer selected from 0, 1, 2, 3 or 4; and
p is an integer selected from 2, 3 or 4.
Compounds of the invention have been shown to be bromodomain inhibitors, in particular BD2 selective and may be useful in the treatment of various diseases or conditions, for example acute or chronic auto-immune and/or inflammatory conditions, for example rheumatoid arthritis and cancer. Accordingly, the invention is further directed to pharmaceutical compositions comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof. The invention is still further directed to methods of treatment of diseases or conditions associated with bromodomains using a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof.
DETAILED DESCRIPTION OF THE INVENTION
Compounds of formula (I) and salts thereof are referred to herein as “compounds of the invention”.
“BD2” refers to Binding Domain 2 of any of the the BET family of proteins BRD2, BRD3, BRD4 or BRDT.
“Alkyl” refers to a saturated hydrocarbon chain having the specified number of carbon atoms. For example, the term “C1-3alkyl” and “C1-4alkyl” as used herein refers to a straight or branched alkyl group having from 1 to 3 or 1 to 4 carbon atoms respectively. Further, the term “C0-3alkyl” refers to a straight or branched alkyl group having from 0 (i.e. a bond) to 3 carbon atoms. Representative branched alkyl groups have one, two or three branches. An alkyl group may form part of a chain, for example, —C0-3alkylOR6 refers to a straight or branched alkyl chain having from 0 (i.e. a bond) to 3 carbon atoms linked to a group R6. “Alkyl” includes, but is not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, t-butyl, pentyl and hexyl.
“Cycloalkyl” refers to a saturated hydrocarbon mono or bicyclic ring or a saturated spiro-linked bicyclic hydrocarbon ring, having 3, 4, 5, 6, 7, 8, 9 or 10 member atoms in the ring. Suitable examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl, spiro[3.3]heptanyl, bicyclo[2.2.1]heptanyl, adamantyl, bicyclo[3.1.0]hexanyl and bicyclo[2.2.2]octanyl. “C3-7cycloalkyl” refers to a saturated hydrocarbon mono or bicyclic ring or a saturated spiro-linked bicyclic hydrocarbon ring, having 3, 4, 5, 6 or 7 member atoms in the ring. Examples of C3-7cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl and bicyclo[3.1.0]hexanyl.
“Halo” refers to a halogen radical, for example, fluoro, chloro, bromo, or iodo.
“Heteroaryl” refers to a monocyclic or bicyclic group having 5, 6, 8, 9, 10 or 11 member atoms, including one, two or three heteroatoms independently selected from nitrogen, sulphur and oxygen, wherein at least a portion of the group is aromatic. The point of attachment to the rest of the molecule may be by any suitable carbon or nitrogen atom. Examples of “heteroaryl” groups include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, benzofuranyl, isobenzofuryl, 2,3-dihydrobenzofuryl, 1,3-benzodioxolyl, dihydrobenzodioxinyl, benzothienyl, benzazepinyl, 2,3,4,5-tetrahydro-1H-benzo[d]azepinyl, indolizinyl, indolyl, indolinyl, isoindolyl, dihydroindolyl, benzimidazolyl, dihydrobenzimidazolyl, benzoxazolyl, dihydrobenzoxazolyl, benzthiazolyl, benzoisothiazolyl, dihydrobenzoisothiazolyl, indazolyl, imidazopyridinyl, pyrazolopyridinyl, pyrrolopyridinyl, benzotriazolyl, triazolopyridinyl, purinyl, quinolinyl, tetrahydroquinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinazolinyl, 1,5-naphthyridinyl, 1,6-naphthyridinyl, 1,7-naphthyridinyl, 1,8-naphthyridinyl, and pteridinyl.
“C5-6 heteroaryl” refers to a monocyclic aromatic group having 5 or 6 member atoms, including 1, 2, 3 or 4 heteroatoms independently selected from nitrogen, sulphur and oxygen. The point of attachment to the rest of the molecule may be by any suitable carbon or nitrogen atom. Examples of “C5-6 heteroaryl” groups include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl, pyridazinyl, pyrazinyl, pyrimidinyl and triazinyl.
“Heteroatom” refers to a nitrogen, sulfur, or oxygen atom.
“Heterocyclyl” refers to a non-aromatic heterocyclic monocyclic or bicyclic ring system containing 4, 5, 6, 7, 8, 9 or 10 ring member atoms, including one heteroatom and optionally containing a further heteroatom selected from nitrogen, oxygen or sulphur. Examples of “heterocyclyl” groups include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolidinyl, pyrazolinyl, imidazolidinyl, imidazolinyl, oxazolinyl, thiazolinyl, tetrahydrofuranyl, dihydrofuranyl, 1,3-dioxolanyl, piperidinyl, piperazinyl, homopiperazinyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-oxathiolanyl, 1,3-oxathianyl, 1,3-dithianyl, 1,4-oxathiolanyl, 1,4-oxathianyl, 1,4-dithianyl, morpholinyl, thiomorpholinyl, hexahydro-1H-1,4-diazepinyl, azabicyclo[3.2.1]octyl, azabicyclo[3.3.1]nonyl, azabicylco[4.3.0]nonyl, oxa bicyclo[2.2.1]heptyl, 1,1-dioxidotetrahydro-2H-thiopyranyl, 1,5,9-triazacyclododecyl, 3-oxabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.1.0]hexanyl, (1r,5s)-3-oxabicyclo[3.1.0]hexanyl and (1r,5s)-3-azabicyclo[3.1.0]hexanyl.
“4 to 7-membered heterocyclyl” refers to a non-aromatic heterocyclic ring system containing 4, 5, 6 or 7 ring member atoms, including one heteroatom and optionally containing a further heteroatom selected from nitrogen, oxygen or sulphur. Examples of “4 to 7-membered heterocyclyl” groups include, but are not limited to, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl.
“Member atoms” refers to the atom or atoms that form a chain or ring. Where more than one member atom is present in a chain and within a ring, each member atom is covalently bound to an adjacent member atom in the chain or ring. Atoms that make up a substituent group attached to a chain or ring are not member atoms in the chain or ring.
“Substituted” in reference to a group indicates that a hydrogen atom attached to a member atom within a group is replaced. It should be understood that the term “substituted” includes the implicit provision that such substitution be in accordance with the permitted valence of the substituted atom and the substituent and that the substitution results in a stable compound (i.e. one that does not spontaneously undergo transformation such as rearrangement, cyclisation, or elimination). In certain embodiments, a single atom may be substituted with more than one substituent as long as such substitution is in accordance with the permitted valence of the atom. Suitable substituents are defined herein for each substituted or optionally substituted group.
“Pharmaceutically acceptable” refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
“Pharmaceutically acceptable excipient” refers to a pharmaceutically acceptable material, composition or vehicle involved in giving form or consistency to the pharmaceutical composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of formula (I) or a pharmaceutically acceptable salt thereof when administered to a patient are avoided. In addition, each excipient must of course be pharmaceutically acceptable e.g. of sufficiently high purity.
“rac” refers to the racemic mixture of the compounds of formula (I).
The compounds of the invention may exist in solid or liquid form. In the solid state, the compounds of the invention may exist in crystalline or non-crystalline form, or as a mixture thereof. For compounds of the invention that are in crystalline form, the skilled artisan will appreciate that pharmaceutically acceptable solvates may be formed wherein solvent molecules are incorporated into the crystalline lattice during crystallization. Solvates may involve non-aqueous solvents such as ethanol, iso-propyl alcohol, dimethylsulfoxide (DMSO), acetic acid, ethanolamine, and ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice. Solvates wherein water is the solvent that is incorporated into the crystalline lattice are typically referred to as “hydrates”. Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The invention includes all such solvates.
It will be further appreciated that certain compounds of the invention that exist in crystalline form, including the various solvates thereof, may exhibit polymorphism (i.e. the capacity to occur in different crystalline structures). These different crystalline forms are typically known as “polymorphs”. The invention includes such polymorphs. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. It will be appreciated that different polymorphs may be produced, for example, by changing or adjusting the reaction conditions or reagents, used in making the compound. For example, changes in temperature, pressure, or solvent may result in polymorphs. In addition, one polymorph may spontaneously convert to another polymorph under certain conditions. Polymorphic forms of compounds of formula (I) may be characterized and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, Raman spectra, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid state nuclear magnetic resonance (SSNMR).
The compounds according to formula (I) may contain one or more asymmetric centres (also referred to as a chiral centres) and may, therefore, exist as individual enantiomers, diastereoisomers, or other stereoisomeric forms, or as mixtures thereof. Chiral centres, such as chiral carbon atoms, may also be present in a substituent such as an alkyl group. Where the stereochemistry of a chiral centre present in formula (I), or in any chemical structure illustrated herein, is not specified, the structure is intended to encompass any stereoisomer and all mixtures thereof. Thus, compounds according to formula (I) containing one or more chiral centres may be used as racemic mixtures, enantiomerically-enriched mixtures, or as enantiomerically-pure individual stereoisomers. Accordingly, the present invention encompasses all isomers of the compounds of formula (I) whether as individual isomers isolated such as to be substantially free of the other isomer (i.e. pure) or as mixtures (i.e. racemic mixtures). An individual isomer isolated such as to be substantially free of the other isomer (i.e. pure) may be isolated such that less than 10%, particularly less than about 1%, for example less than about 0.1% of the other isomer is present.
Racemic compounds with a single stereocentre are denoted with either no stereochemistry (single bond) or have the annotation (+/−) or rac. Racemic compounds with two or more stereocentres where relative stereochemistry is known are denoted cis or transas drawn in the structure. Resolved single enantiomers with unknown absolute stereochemistry but known relative stereochemistry are referred to with (R* or S*) with the appropriate relative stereochemistry depicted.
Where diastereoisomers are represented and only the relative stereochemistry is referred to, the bold or hashed solid bond symbols (
Figure US10966961-20210406-P00001
/
Figure US10966961-20210406-P00002
) are used. Where the absolute stereochemistry is known and the compound is a single enantiomer, the bold or hashed wedges symbols (
Figure US10966961-20210406-P00003
/
Figure US10966961-20210406-P00004
) are used as appropriate.
Individual stereoisomers of a compound according to formula (I) which contain one or more asymmetric centres may be resolved by methods known to those skilled in the art. For example, such resolution may be carried out (1) by formation of diastereoisomeric salts, complexes or other derivatives; (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where the desired stereoisomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired form. Alternatively, specific stereoisomers may be synthesised by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
It will be appreciated that, for compounds of formula (I) tautomers may be observed. Any comment relating to the biological activity of a tautomer should be taken to include both tautomers. It is to be understood that the references herein to compounds of formula (I) and salts thereof covers the compounds of formula (I) as free bases, or as salts thereof, for example as pharmaceutically acceptable salts thereof. Thus, in one embodiment, the invention is directed to compounds of formula (I) as the free base. In another embodiment, the invention is directed to compounds of formula (I) and salts thereof. In a further embodiment, the invention is directed to compounds of formula (I) and pharmaceutically acceptable salts thereof.
Because of their potential use in medicine, salts of the compounds of formula (I) are desirably pharmaceutically acceptable. Suitable pharmaceutically acceptable salts can include acid addition salts or base addition salts. For a review of suitable pharmaceutically acceptable salts see Berge et al., J. Pharm. Sci., 66:1-19, (1977). Typically, a pharmaceutically acceptable salt may be readily prepared by using a desired acid or base as appropriate. The resultant salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
A pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinic, maleic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, aspartic, p-toluenesulphonic, benzenesulphonic, methanesulphonic, ethanesulphonic, naphthalenesulphonic such as 2-naphthalenesulphonic, or hexanoic acid), optionally in a suitable solvent such as an organic solvent, to give the salt which is usually isolated for example by crystallisation and filtration or by evaporation followed by trituration. A pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, succinate, maleate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulphonate, benzenesulphonate, methanesulphonate, ethanesulphonate, naphthalenesulphonate (e.g. 2-naphthalenesulphonate) or hexanoate salt.
Other non-pharmaceutically acceptable salts, e.g. formates or trifluoroacetates, may be used, for example in the isolation of the compounds of formula (I), and are included within the scope of this invention.
The invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compounds of formula (I).
It will be appreciated from the foregoing that included within the scope of the invention are solvates, isomers and polymorphic forms of the compounds of formula (I) and salts thereof.
Statement of the Invention
In a first aspect there are provided compounds of formula (I):
Figure US10966961-20210406-C00002

or a salt thereof
wherein:
R1 is —C1-3alkyl or cyclopropyl;
R2 is —C0-3alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R5 groups which may be the same or different; or
R2 is —C0-4alkyl-heterocyclyl or —(CH2)pO-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different; or
R2 is H, —CH3, —C2-6alkyl optionally substituted by one, two, three, four or five fluoro, —C2-6alkyl OR6, —C2-6alkyl NR10aR11a, —(CH2)mSO2C1-3alkyl, —(CH2)mSO2NR10R11, —(CH2)mC(O)NR10R11, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mNHCO2C1-4alkyl, —(CH2)mNHC(O)C1-4alkyl or —(CH2)nheteroaryl wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different;
R3 is H, —C1-4alkyl, cyclopropyl, —CH2F, —C1-3alkylOR6 or —C1-3alkylCN;
R4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R7 groups which may be the same or different;
each R5 is independently halo, —C0-6alkyl-R8, —O—C2-6alkyl-R8, —O—CH2phenyl, —CN or —SO2C1-3alkyl;
R6 is H or —C1-4alkyl;
each R7 is independently oxo, halo, —C1-4alkyl optionally substituted by one, two or three fluoro, —C0-3alkylOR6, —OC2-3alkylOR6, —C0-3alkylNR10R11, —C0-3alkyl-CONR10R11, —CN, —SO2—C1-3alkyl, —SO2NR10R11 or —SO2phenyl optionally substituted by —C1-4alkyl
R8 is H, —OR6, —NR10R11 or heteroaryl;
each R9 is independently halo, —C1-4alkyl, cyclopropyl, cyclobutyl, —CH2CF3, —CH2CHF2, —CH2CH2F, —OCH2CH2OR6, —C0-3alkylOR6, —C0-3alkylNR10R11, —NHCH2CH2OR6, —NHCO2C1-4alkyl, oxo, —C(O)R6, —C(O)OR6 or —C(O)NR10R11;
R10 and R11 are each independently selected from H and —C1-3alkyl; or R10 and R11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C1-3alkyl optionally substituted with one, two or three fluorine atoms, —C2-4alkylOH, —OH and F;
R10a and R11a are each independently selected from H and —C1-3alkyl;
m is an integer selected from 2, 3 or 4;
n is an integer selected from 0, 1, 2, 3 or 4; and
p is an integer selected from 2, 3 or 4.
In one embodiment there is provided a compound of formula (I) or a salt thereof wherein:
R1 is —C1-3alkyl or cyclopropyl;
R2 is —C0-3alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R5 groups which may be the same or different; or
R2 is —C0-4alkyl-heterocyclyl or —(CH2)pO-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different; or
R2 is H, —CH3, —C2-6alkyl optionally substituted by up to five fluoro, —C2-6alkylOR6, —C2-6alkylNR10aR11a, —(CH2)mSO2C1-3alkyl, —(CH2)mSO2NR10R11, —(CH2)mC(O)NR10R11, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mNHCO2C1-4alkyl, —(CH2)mNHC(O)C1-4alkyl or —(CH2)nheteroaryl wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different;
R3 is —H, —C1-4alkyl, cyclopropyl, —CH2F, —C1-3alkylOR6 or —C1-3alkylCN;
R4 is phenyl or a heteroaryl group wherein each are optionally substituted by one, two or three R7 groups which may be the same or different;
each R5 is independently halo, —C0-6alkyl-R8, —O—C2-6alkyl-R8, —CN or —SO2C1-3alkyl; R6 is H or —C1-4alkyl;
each R7 is independently oxo, halo, —C1-4alkyl optionally substituted by 1, 2 or 3 fluoro, —C0-3alkylOR6, —OC0-3alkylOR6, —C0-3alkylNR10R11, —C0-3alkyl-CONR10R11, —CN, —SO2—C1-3alkyl or —SO2NR10R11;
R8 is H, —OR6, —NR10R11 or heteroaryl;
each R9 is independently halo, —C1-4alkyl, cyclopropyl, cyclobutyl, —CH2CF3, —CH2CHF2, —CH2CH2F, —OCH2CH2OR6, —C0-3alkylOR6, —C0-3alkylNR10R11, —NHCH2CH2OR6, —NHCO2R6, oxo, —C(O)R6, —C(O)OR6 or —C(O)NR10R11;
R10 and R11 are each independently selected from —H and —C1-3alkyl; or R10 and R11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C1-3alkyl optionally substituted with up to 3 fluorine atoms, —C2-4alkylOH, —OH and F;
R10a and R11a are each independently selected from —H and —C1-3alkyl;
m is an integer selected from 2, 3 or 4;
n is an integer selected from 0, 1, 2, 3 or 4; and
p is an integer selected from 2, 3 or 4.
In one embodiment R1 is methyl.
In one embodiment R2 is —C0-3alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two or three R5 groups which may be the same or different. In one embodiment R2 is a —C0-3alkyl-C3-7cycloalkyl group, wherein the C3-7cycloalkyl group is selected from the group consisting of cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl, said groups being optionally substituted with one, two or three R5 groups which may be the same or different. In another embodiment R2 is cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl optionally substituted with one, two or three R5 groups which may be the same or different. In another embodiment R2 is cyclopropyl, cyclobutyl, cyclohexyl or bicyclo[3.1.0]hexanyl optionally substituted with one R5 group selected from methyl, fluoro and —OH. In a particular embodiment R2 is a cyclopropyl optionally substituted by one methyl group. In another particular embodiment R2 is a cyclohexyl group optionally substituted with a OH group. In another embodiment R2 is a cyclohexyl group optionally substituted with a methoxy group.
In one embodiment R2 is —C0-4alkyl-heterocyclyl or —(CH2)pO-heterocyclyl wherein each heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different. In another embodiment R2 is —C0-4alkyl-heterocyclyl wherein the heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different. In another embodiment R2 is —C0-4alkyl-heterocyclyl which is -heterocyclyl, —CH2CH2-heterocyclyl or —CH2CH2CH2-heterocyclyl. In another embodiment R2 is —C0-4alkyl-heterocyclyl wherein the heterocyclyl is selected from oxetanyl, tetrahydrofuranyl, tetrahydro-2/pyranyl, morpholinyl, piperidinyl, piperazinyl, (1 r,5s)-3-oxa bicyclo[3.1.0]hexanyl and (1r,5S)-3-azabicyclo[3.1.0]hexanyl said groups being optionally substituted by one or two R9 groups which may be the same or different. In another embodiment R2 is —C0-4alkyl-heterocyclyl wherein the heterocyclyl is selected from oxetanyl, tetrahydrofuranyl, tetrahydro-2/pyranyl, morpholinyl, piperidinyl, piperazinyl, (1r,5S)-3-oxabicyclo[3.1.0]hexanyl and (1r,5s)-3-azabicyclo[3.1.0]hexanyl optionally substituted by one or two R9 groups selected from methyl, —C(O)CH3 and fluoro. In a further embodiment R2 is —C0-4alkyl-heterocyclyl wherein the heterocyclyl optionally substituted by one or two R9 groups, is selected from:
Figure US10966961-20210406-C00003
In another embodiment R2 is H, —CH3, C2-6alkyl optionally substituted by one, two, three, four or five fluoro, —C2-6alkylOR6, —C2-6alkylNR19R11, —(CH2)mSO2C1-3alkyl, —(CH2)mSO2NR19R11, —(CH2)mC(O)NR19R11, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mNHCO2C1-4alkyl —(CH2)mNHC(O)C1-4alkyl or —(CH2)nheteroaryl wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different.
In another embodiment R2 is selected from methyl, ethyl, propyl, iso-propyl, butyl, —CH2CH2CH(CH3)2, —CH2CH(CH3)2, —CH2CH2OR6, —CH2CH2CH2OR6, —CH2CH(CH3)OR6, —CH2CH2CH(CH3)OR6—CH2CH2CH(CH3)NR10R11, —CH2CH2CH2NR10R11, —(CH2)mSO2CH3, —(CH2)mC(O)NHCH3, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mCF3 and —(CH2)mNHCO2C(CH3)3.
In another embodiment R2 is —C1-6alkyl selected from methyl, ethyl, propyl, iso-propyl, butyl, —CH2CH2CH(CH3)2 and —CH2CH(CH3)2. In another embodiment R2 is —C1-6alkylOR6 selected from —CH2CH2OR6, —CH2CH2CH2OR6, —CH2CH(CH3)OR6 and —CH2CH2CH(CH3)OR6. In another embodiment R2 is —C 1-6alkylNR10R11 selected from —CH2CH2CH(CH3)NR10R11 and —CH2CH2CH2NR10R11. In another embodiment R2 is —(CH2)mSO2CH3. In another embodiment R2 is —(CH2)mC(O)NHCH3. In another embodiment R2 is —(CH2)mCN. In another embodiment R2 is —(CH2)mCO2R6. In another embodiment R2 is —(CH2)mCF3. In another embodiment R2 is —(CH2)mNHCO2C(CH3)3.
In one embodiment R2 is —(CH2)nheteroaryl wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different. In a further embodiment R2 is —(CH2)nC5-6heteroaryl wherein the C5-6heteroaryl is selected from furanyl, thienyl, pyrrolyl, triazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridinyl, pyridazinyl, pyrazinyl and pyrimidinyl said groups being optionally substituted by one or two R′ substituents independently selected from halo, C1-4alkyl (such as methyl) and —C0-3alkylOR6. In another embodiment there is provided compounds of formula (I) in which R2 is —(CH2)nC5-6heteroaryl wherein the C5-6heteroaryl is pyrazolyl optionally substituted by C1-4alkyl or —C0-3alkylOR6. In a particular embodiment there is provided compounds of formula (I) in which R2 is —(CH2)nC5-6heteroaryl wherein the optionally substituted C5-6heteroaryl group is selected from the group consisting of
Figure US10966961-20210406-C00004

wherein * denotes the point of attachment.
In one embodiment R3 is H, methyl, ethyl, —CH2F, —CH2OH, —CH(OH)CH3, —CH2OMe or —CH2CN. In one embodiment R3 is H, methyl, ethyl or CH2OH.
In one embodiment R4 is phenyl optionally substituted by one, two or three R7 groups which may be the same or different. In another embodiment R4 is unsubstituted phenyl. In another embodiment R4 is phenyl substituted by one or two R7 groups which may be the same or different selected from halo, —C1-4alkyl optionally substituted one, two or three fluoro, —C0-3alkylOR6, —OC2-3alkylOR6 and CN. In another embodiment R4 is phenyl substituted by one or two R7 groups which may be the same or different selected from halo, —C1-4alkyl, —C0-3alkylOR6 and CN. In another embodiment R4 is phenyl substituted by one R7 groups selected from the group consisting of fluoro, chloro, methyl, cyano and methoxy.
In another embodiment R4 is a heteroaryl group which is pyridyl optionally substituted by one, two or three R7 groups which may be the same or different. In another embodiment R4 is a heteroaryl group which is unsubstituted pyridyl.
In another embodiment R4 is a heteroaryl group which is indolyl (e.g 1H-indol-4-yl or 1H-indol-5-yl) optionally substituted by one, two or three R7 groups which may be the same or different. In another embodiment R4 is a heteroaryl group which is 1H-indol-4-yl.
In another embodiment R4 is a heteroaryl group which is a pyrrolopyridinyl (e.g. 1H-pyrrolo[2,3,b]pyridinyl or 1H-pyrrolo[2,3,c]pyridinyl)) optionally substituted by one, two or three R7 groups which may be the same or different. In another embodiment R4 is a heteroaryl group which is unsubstituted pyrrolopyridinyl.
In one embodiment each R5 is independently halo or —C0-6alkyl-R8 wherein R8 is H, OR6 (such as OH) or NR10R11 (such as NH2).
In one embodiment R6 is H, methyl, ethyl or t-butyl.
In one embodiment each R7 is independently oxo, halo, —C1-4alkyl optionally substituted by one, two or three fluoro, —C0-3alkylOR6, —C0-3alkylNR10R11, —C0-3alkyl-CONR10R11, —CN, —SO2—C1-3alkyl or —SO2NR10R11. In another embodiment each R7 is independently halo, —C1-4alkyl optionally substituted by one, two or three fluoro, —C0-3alkylOR6 or CN.
In one embodiment each R7 is independently halo, —C1-4alkyl, —C0-3alkylOR6 or CN; In one embodiment R8 is H, —OH or methoxy.
In one embodiment each R9 is independently halo, C1-4alkyl (such as methyl), —C0-3alkylOR6, —C0-3alkylNR10R11, oxo, or —C(O)R6 (such as C(O)CH3).
In one embodiment m is 2 or 3.
In one embodiment n is 0, 1 or 2. In a further embodiment n is 0. In a yet further embodiment n is 2.
In one embodiment p is 2 or 3.
It is to be understood that the present invention covers all combinations of substituent groups described hereinabove.
Compounds of the invention include the compounds of Examples 1 to 261 and salts thereof.
Compounds of the invention include the compounds of Examples 1 to 138 and salts thereof.
In one embodiment the compound of formula (I) is selected from:
  • N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide;
  • N5-((1r,4S)-4-hydroxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide; and
  • N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
    • or a salt thereof.
In one embodiment there is provided
Figure US10966961-20210406-C00005

or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided
Figure US10966961-20210406-C00006
In one embodiment there is provided
Figure US10966961-20210406-C00007

or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided
Figure US10966961-20210406-C00008
In another embodiment the compound of formula (I) is selected from:
  • N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-((S)-1-(4-chlorophenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide;
  • (S)—N5-methyl-1-(1-phenylethyl)-N5-(1H-pyrazol-4-yl)-1H-pyrazole-3,5-dicarboxamide;
  • 1-((S)-1-(4-chlorophenyl)ethyl)-N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide;
  • 1-((S)-1-(3-chlorophenyl)ethyl)-N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide; and
  • N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylpropyl)-1H pyrazole-3,5-dicarboxamide
    or a salt thereof.
In a second aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
In a third aspect of the present invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof for use in therapy, in particular in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
In a fourth aspect of the present invention, there is provided a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
In a fifth aspect of the present invention, there is provided the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
The compounds of the invention may possess an improved profile over known BET inhibitors (including properties such as potency, selectivity and/or developability). Certain compounds of the invention may have an advantageous combination of such properties.
Statement of Use
The compounds of formula (I) and salts thereof are bromodomain inhibitors, and thus are believed to have potential utility in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
Bromodomain inhibitors are believed to be useful in the treatment of a variety of diseases or conditions related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the prevention and treatment of viral infections.
Bromodomain inhibitors may be useful in the treatment of a wide variety of acute or chronic autoimmune and/or inflammatory conditions such as rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease (Crohn's disease and ulcerative colitis), asthma, chronic obstructive airways disease, pneumonitis, myocarditis, pericarditis, myositis, eczema, dermatitis (including atopic dermatitis), alopecia, vitiligo, bullous skin diseases, nephritis, vasculitis, hypercholesterolennia, atherosclerosis, Alzheimer's disease, Sjögren's syndrome, sialoadenitis, central retinal vein occlusion, branched retinal vein occlusion, Irvine-Gass syndrome (post cataract and post-surgical), retinitis pigmentosa, pars planitis, birdshot retinochoroidopathy, epiretinal membrane, cystic macular edema, parafoveal telengiectasis, tractional maculopathies, vitreomacular traction syndromes, retinal detachment, neuroretinitis, idiopathic macular edema, retinitis, dry eye (keratoconjunctivitis Sicca), vernal keratoconjunctivitis, atopic keratoconjunctivitis, uveitis (such as anterior uveitis, pan uveitis, posterior uveitis, uveitis-associated macular edema), scleritis, diabetic retinopathy, diabetic macula edema, age-related macular dystrophy, hepatitis, pancreatitis, primary biliary cirrhosis, sclerosing cholangitis, acute alcoholic hepatitis, chronic alcoholic hepatitis, alcoholic steato-hepatitis, non-alcoholic steato-hepatitis (NASH), cirrhosis, Childs-Pugh cirrhosis, autoimmune hepatitis, fulminant hepatitis, chronic viral hepatitis, alcoholic liver disease, systemic sclerosis, systemic sclerosis with associated interstitial lung disease, sarcoidosis, neurosarcoidosis, Addison's disease, hypophysitis, thyroiditis, Type I diabetes, Type II diabetes, giant cell arteritis, nephritis including lupus nephritis, vasculitis with organ involvement such as glomerulonephritis, vasculitis including giant cell arteritis, Wegener's granulomatosis, Polyarteritis nodosa, Behcet's disease, Kawasaki disease, Takayasu's Arteritis, pyoderma gangrenosum, vasculitis with organ involvement, acute rejection of transplanted organs and systemic sclerosis.
In one embodiment the acute or chronic autoimmune and/or inflammatory condition is a disorder of lipid metabolism mediated via the regulation of APO-A1 such as hypercholesterolemia, atherosclerosis or Alzheimer's disease.
In another embodiment the acute or chronic autoimmune and/or inflammatory condition is a respiratory disorder such as asthma or chronic obstructive airways disease.
In another embodiment the acute or chronic autoimmune and/or inflammatory condition is a systemic inflammatory disorder such as rheumatoid arthritis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis or inflammatory bowel disease (Crohn's disease or Ulcerative colitis).
In another embodiment, the acute or chronic autoimmune and/or inflammatory condition is multiple sclerosis.
In another embodiment, the acute or chronic autoimmune and/or inflammatory condition is Type I diabetes.
In another embodiment, the acute or chronic autoimmune and/or inflammatory condition is rheumatoid arthritis.
Bromodomain inhibitors may be useful in the treatment of depression.
Bromodomain inhibitors may be useful in the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins, such as sepsis, acute sepsis, sepsis syndrome, septic shock, endotoxaemia, systemic inflammatory response syndrome (SIRS), multi-organ dysfunction syndrome, toxic shock syndrome, acute lung injury, ARDS (adult respiratory distress syndrome), acute renal failure, fulminant hepatitis, burns, acute pancreatitis, post-surgical syndromes, sarcoidosis, Herxheimer reactions, encephalitis, myelitis, meningitis, malaria and SIRS associated with viral infections such as influenza, herpes zoster, herpes simplex and coronavirus. In one embodiment the disease or condition which involves an inflammatory response to an infection with bacteria, a virus, fungi, a parasite or their toxins is acute sepsis.
Bromodomain inhibitors may be useful in the treatment of conditions associated with ischaemia-reperfusion injury such as myocardial infarction, cerebro-vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
Bromodomain inhibitors may be useful in the treatment of cardiovascular diseases such as coronary artery diseases (for example, angina or myocardial infarction), pulmonary arterial hypertension, cerebro-vascular ischaemia (stroke), hypertensive heart disease, rheumatic heart disease, cardiomyopathy, atrial fibrillation, congenital heart disease, endocarditis, aortic aneurysms or peripheral artery disease.
Bromodomain inhibitors may be useful in the treatment of fibrotic conditions such as idiopathic pulmonary fibrosis, pulmonary fibrosis, cystic fibrosis, progressive massive fibrosis, renal fibrosis, liver fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), post-operative stricture, keloid scar formation, scleroderma (including morphea and systemic sclerosis), cardiac fibrosis, atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, arthrofibrosis, Dupuytren's contracture, mediastinal, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis and adhesive capsulitis.
Bromodomain inhibitors may be useful in the treatment of viral infections such as herpes simplex infections and reactivations, cold sores, herpes zoster infections and reactivations, chickenpox, shingles, human papilloma virus (HPV), human immunodeficiency virus (HIV), cervical neoplasia, adenovirus infections, including acute respiratory disease, poxvirus infections such as cowpox or smallpox, or African swine fever virus. In one embodiment the viral infection is a HPV infection of skin or cervical epithelia. In another embodiment the viral infection is a latent HIV infection.
Bromodomain inhibitors may be useful in the treatment of a wide variety of bone disorders such as osteoporosis, osteopenia, osteoarthritis and ankylosing spondylitis.
Bromodomain inhibitors may be useful in the treatment of cancer, including hematological cancers (such as leukaemia, lymphoma and multiple myeloma), epithelial cancers (including lung, breast or colon carcinomas), midline carcinomas, or mesenchymal, hepatic, renal or neurological tumours.
Bromodomain inhibitors may be useful in the treatment of one or more cancers selected from brain cancer (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast cancer, inflammatory breast cancer, colorectal cancer, Wilm's tumor, Ewing's sarcoma, rhabdomyosarcoma, ependymoma, medulloblastoma, colon cancer, head and neck cancer, kidney cancer, lung cancer, liver cancer, melanoma, squamous cell carcinoma, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma cancer, osteosarcoma, giant cell tumor of bone, thyroid cancer, lymphoblastic T-cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T-cell leukemia, plasmacytoma, immunoblastic large cell leukemia, mantle cell leukemia, multiple myeloma, megakaryoblastic leukemia, acute megakaryocytic leukemia, promyelocytic leukemia, mixed lineage leukaemia, erythroleukemia, malignant lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, lymphoblastic T-cell lymphoma, Burkitt's lymphoma, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), NUT-midline carcinoma and testicular cancer.
In one embodiment the cancer is a leukaemia, for example a leukaemia selected from acute monocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia and mixed lineage leukaemia (MLL). In another embodiment the cancer is NUT-midline carcinoma. In another embodiment the cancer is multiple myeloma. In another embodiment the cancer is a lung cancer such as small cell lung cancer (SCLC). In another embodiment the cancer is a neuroblastoma. In another embodiment the cancer is Burkitt's lymphoma. In another embodiment the cancer is cervical cancer. In another embodiment the cancer is esophageal cancer. In another embodiment the cancer is ovarian cancer. In another embodiment the cancer is breast cancer. In another embodiment the cancer is colorectal cancer. In another embodiment the cancer is prostate cancer. In another embodiment the cancer is castration resistant prostate cancer.
Bromodomain inhibitors may be useful in the treatment of diseases associated with systemic inflammatory response syndrome, such as sepsis, burns, pancreatitis, major trauma, haemorrhage and ischaemia. In this embodiment, the bromodomain inhibitor would be administered at the point of diagnosis to reduce the incidence of: SIRS, the onset of shock, multi-organ dysfunction syndrome, which includes the onset of acute lung injury, ARDS, acute renal, hepatic, cardiac or gastro-intestinal injury and mortality. In another embodiment the bromodomain inhibitor would be administered prior to surgical or other procedures associated with a high risk of sepsis, haemorrhage, extensive tissue damage, SIRS or MODS (multiple organ dysfunction syndrome). In a particular embodiment the disease or condition for which a bromodomain inhibitor is indicated is sepsis, sepsis syndrome, septic shock and endotoxaemia. In another embodiment, the bromodomain inhibitor is indicated for the treatment of acute or chronic pancreatitis. In another embodiment the bromodomain is indicated for the treatment of burns.
The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy. In one embodiment there is provided N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide, or a pharmaceutically salt thereof for use in therapy.
The compound of formula (I) or a pharmaceutically salt thereof can be used in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of a disease or condition for which a bromodomain inhibitor is indicated. In one embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of acute or chronic auto-immune and/or inflammatory conditions. In one embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of rheumatoid arthritis. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of conditions associated with ischaemia-reperfusion injury. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cardiovascular diseases. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of fibrotic conditions. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of viral infections. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of bone disorders. In another embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cancer. In a further embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases associated with systemic inflammatory response syndrome.
Also provided is the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. In one embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of acute or chronic auto-immune and/or inflammatory conditions. In one embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of rheumatoid arthritis. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of conditions associated with ischaemia-reperfusion injury. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cardiovascular diseases. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of fibrotic conditions. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of viral infections. In another embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cancer. In a further embodiment there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases associated with systemic inflammatory response syndrome.
Also provided is a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof. In one embodiment there is provided a method of treating acute or chronic auto-immune and/or inflammatory conditions in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In one embodiment there is provided a method of treating rheumatoid arthritis in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating conditions associated with ischaemia-reperfusion injury in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating cardiovascular diseases in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating fibrotic conditions in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating viral infections in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a method of treating cancer in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In a further embodiment there is provided a method of treating diseases associated with systemic inflammatory response syndrome in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
Suitably the subject in need thereof is a mammal, particularly a human.
The invention further provides for a method for inhibiting a bromodomain containing protein which comprises contacting the bromodomain containing protein with a compound of formula (I) or a pharmaceutically acceptable salt thereof.
As used herein the reference to the “treatment” of a particular disease or condition includes the prevention or prophylaxis of such a disease or condition.
Pharmaceutical Compositions/Routes of Administration/Dosages
Compositions
While it is possible that for use in therapy, a compound of formula (I) as well as pharmaceutically acceptable salts thereof may be administered as the raw chemical, it is common to present the active ingredient as a pharmaceutical composition. The compounds of formula (I) and pharmaceutically acceptable salts thereof will normally, but not necessarily, be formulated into pharmaceutical compositions prior to administration to a patient. Accordingly, in another aspect there is provided a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more (e.g. two, three, four, five or six) pharmaceutically acceptable excipients. In one embodiment there is provided N5-(1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients. The compounds of formula (I) and pharmaceutically acceptable salts are as described above. The excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical composition including admixing a compound of formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable excipients. A pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof may be prepared by, for example, admixture at ambient temperature and atmospheric pressure. The pharmaceutical composition can be used in the treatment of any of the conditions described herein.
In a further aspect the invention is directed to pharmaceutical compositions for the treatment of a disease or condition for which a bromodomain inhibitor is indicated comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof.
Since the compounds of formula (I) are intended for use in pharmaceutical compositions it will be readily understood that they are each preferably provided in substantially pure form, for example, at least 85% pure, especially at least 98% pure (% in a weight for weight basis).
Pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered more than once a day. Preferred unit dosage compositions are those containing a daily dose or sub-dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient.
Pharmaceutical compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), ocular (including topical, intraocular, subconjunctival, episcleral, sub-Tenon), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
The pharmaceutical compositions of the invention may be prepared and packaged in bulk form wherein a safe and effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof can be extracted and then given to the patient such as with powders or syrups. Alternatively, the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form wherein each physically discrete unit contains a compound of formula (I) or a pharmaceutically acceptable salt thereof. When prepared in unit dosage form, the pharmaceutical compositions of the invention typically may contain, for example, from 0.25 mg to 1 g, or from 0.5 mg to 500 mg, or from 1 mg to 100 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
The compound of formula (I) or a pharmaceutically acceptable salt thereof and the pharmaceutically acceptable excipient or excipients will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration. For example, dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixers, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols, solutions, and dry powders; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition. For example, certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting of the compound or compounds of formula (I) or pharmaceutically acceptable salts thereof once administered to the subject from one organ, or portion of the body, to another organ, or portion of the body. Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance subject compliance.
Suitable pharmaceutically-acceptable excipients include the following types of excipients: carriers, diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavouring agents, flavour-masking agents, colouring agents, anti-caking agents, humectants, chelating agents, plasticisers, viscosity increasing agents, antioxidants, preservatives, stabilisers, surfactants, and buffering agents. The skilled artisan will appreciate that certain pharmaceutically-acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other excipients are present in the formulation.
Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically-acceptable excipients and may be useful in selecting suitable pharmaceutically-acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
In one embodiment the pharmaceutical composition is adapted for parenteral administration, particularly intravenous administration.
In one embodiment the pharmaceutical composition is adapted for oral administration.
In one embodiment the pharmaceutical composition is adapted for topical administration.
Pharmaceutical compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions (which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient) and aqueous and non-aqueous sterile suspensions (which may include suspending agents and thickening agents). The compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
Pharmaceutical compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders suitable for incorporating into tablets or capsules may be prepared by reducing the compound to a suitable fine size (e.g. by micronisation) and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
Capsules may be made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
Moreover, when desired or necessary, suitable binders, glidants, lubricants, sweetening agents, flavours, disintegrating agents (disintegrants) and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrants include starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of formula (I) and pharmaceutically acceptable salts thereof can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
Compositions for oral administration may be designed to provide a modified release profile so as to sustain or otherwise control the release of the therapeutically active agent.
Where appropriate, dosage unit compositions for oral administration can be microencapsulated. The composition may be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
For compositions suitable and/or adapted for oral administration, the compound of formula (I) or a pharmaceutically acceptable salt thereof, may be in a particle-size-reduced form e.g. obtained by micronisation. The preferable particle size of the size-reduced (e.g. micronised) compound or salt is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
The compounds of formula (I) and pharmaceutically acceptable salts thereof, can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, emulsions, lotions, powders, solutions, pastes, gels, foams, sprays, aerosols or oils. Such pharmaceutical compositions may include conventional additives which include, but are not limited to, preservatives, solvents to assist drug penetration, co-solvents, emollients, propellants, viscosity modifying agents (gelling agents), surfactants and carriers. In one embodiment there is provided a pharmaceutical composition adapted for topical administration which comprises between 0.01-10%, or between 0.01-1% of the compound of formula (I), or a pharmaceutically acceptable salt thereof, by weight of the composition.
For treatments of the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment, cream, gel, spray or foam. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent. Compositions to be administered to the eye will have ophthalmically compatible pH and osmolality. One or more ophthalmically acceptable pH adjusting agents and/or buffering agents can be included in a composition of the invention, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, and sodium lactate; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases, and buffers can be included in an amount required to maintain pH of the composition in an ophthalmically acceptable range. One or more ophthalmically acceptable salts can be included in the composition in an amount sufficient to bring osmolality of the composition into an ophthalmically acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions.
The ocular delivery device may be designed for the controlled release of one or more therapeutic agents with multiple defined release rates and sustained dose kinetics and permeability. Controlled release may be obtained through the design of polymeric matrices incorporating different choices and properties of biodegradable/bioerodable polymers (e.g. poly(ethylene vinyl) acetate (EVA), superhydrolyzed PVA), hydroxyalkyl cellulose (HPC), methylcellulose (MC), hydroxypropyl methyl cellulose (HPMC), polycaprolactone, poly(glycolic) acid, poly(lactic) acid, polyanhydride, of polymer molecular weights, polymer crystallinity, copolymer ratios, processing conditions, surface finish, geometry, excipient addition and polymeric coatings that will enhance drug diffusion, erosion, dissolution and osmosis.
Pharmaceutical compositions for ocular delivery also include in situ gellable aqueous composition. Such a composition comprises a gelling agent in a concentration effective to promote gelling upon contact with the eye or with lacrimal fluid. Suitable gelling agents include but are not limited to thermosetting polymers. The term “in situgellable” as used herein is includes not only liquids of low viscosity that form gels upon contact with the eye or with lacrimal fluid, but also includes more viscous liquids such as semi-fluid and thixotropic gels that exhibit substantially increased viscosity or gel stiffness upon administration to the eye. See, for example, Ludwig (2005) Adv. Drug Deliv. Rev. 3; 57:1595-639, herein incorporated by reference for purposes of its teachings of examples of polymers for use in ocular drug delivery.
Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions, gels or dry powders.
For compositions suitable and/or adapted for inhaled administration, it is preferred that the compound of formula (I) or a pharmaceutically acceptable salt thereof, is in a particle-size-reduced form e.g. obtained by micronisation. The preferable particle size of the size-reduced (e.g. micronised) compound or salt is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
Aerosol formulations, e.g. for inhaled administration, can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non-aqueous solvent.
Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
Where the dosage form comprises an aerosol dispenser, it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC). Suitable HFC propellants include 1,1,1,2,3,3,3-heptafluoropropane and 1,1,1,2-tetrafluoroethane. The aerosol dosage forms can also take the form of a pump-atomiser. The pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol.
For pharmaceutical compositions suitable and/or adapted for inhaled administration, the pharmaceutical composition may be a dry powder inhalable composition. Such a composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of formula (I) or a pharmaceutically acceptable salt thereof (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid and/or metal salt of stearic acid such as magnesium or calcium stearate. Preferably, the dry powder inhalable composition comprises a dry powder blend of lactose e.g. lactose monohydrate and the compound of formula (I) or salt thereof. Such compositions can be administered to the patient using a suitable device such as the DISKUS® device, marketed by GlaxoSmithKline which is for example described in GB 2242134 A.
The compounds of formula (I) and pharmaceutically acceptable salts thereof may be formulated as a fluid formulation for delivery from a fluid dispenser, for example a fluid dispenser having a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations. The dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity. A fluid dispenser of the aforementioned type is described and illustrated in International Patent Application Publication No. WO 2005/044354 A1.
A therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, will depend upon a number of factors including, for example, the age and weight of the patient, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. In the pharmaceutical composition, each dosage unit for oral or parenteral administration preferably contains from 0.01 mg to 3000 mg, more preferably 0.5 mg to 1000 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base. Each dosage unit for nasal or inhaled administration preferably contains from 0.001 mg to 50 mg, more preferably 0.01 mg to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
The pharmaceutically acceptable compounds of formula (I) and pharmaceutically acceptable salts thereof, can be administered in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day, 0.5 mg to 1000 mg per day or 100 mg to 2500 mg per day, or a nasal or inhaled dose of 0.001 mg to 50 mg per day or 0.01 mg to 5 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base. This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same. An effective amount of a salt thereof, may be determined as a proportion of the effective amount of the compound of formula (I) per se.
The compounds of formula (I) and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents. Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and the use of at least one other therapeutically active agent. The compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. The amounts of the compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. Thus in a further aspect, there is provided a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents.
Thus in one aspect, the compound of formula (I) or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, according to the invention may be used in combination with or include one or more other therapeutic agents, for example selected from antibiotics, anti-virals, glucocorticosteroids, muscarinic antagonists, beta-2 agonists and Vitamin D3 analogues. In a further embodiment a compound of formula (I) or a pharmaceutically acceptable salt thereof may be used in combination with a further therapeutic agent which is suitable for the treatment of cancer. Examples of such further therapeutic agents are described in Cancer Principles and Practice of Oncology by V. T. Devita and S. Hellman (editors), 6th edition (2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Further therapeutic agents to be used in combination with the compound of formula (I) or a pharmaceutically acceptable salt thereof include, but are not limited to, anti-microtubule agents (such as diterpenoids and vinca alkaloids); platinum coordination complexes; alkylating agents (such as nitrogen mustards, oxazaphosphorines, alkylsulphonates, nitrosoureas, and triazenes); antibiotic agents (such as anthracyclins, actinomycins and bleomycins); topoisomerase II inhibitors (such as epipodophyllotoxins); antimetabolites (such as purine and pyrimidine analogues and anti-folate compounds); topoisomerase I inhibitors (such as camptothecins; hormones and hormonal analogues); signal transduction pathway inhibitors (such as tyropsine receptor inhibitors); non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents (such as PD-1 inhibitors, including nivolumab and pembrolizumab, and CTLA-4 inhibitors, including ipilimumab); proapoptotic agents; epigenetic or transcriptional modulators (such as histone deacetylase inhibitors) and cell cycle signaling inhibitors.
It will be appreciated that when the compound of formula (I) or a pharmaceutically acceptable salt thereof, is administered in combination with other therapeutic agents normally administered by the inhaled, intravenous, oral or intranasal route, that the resultant pharmaceutical composition may be administered by the same routes. Alternatively the individual components of the composition may be administered by different routes.
It will be clear to a person skilled in the art that, where appropriate, the other therapeutic agent(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic agent. It will be clear also that, where appropriate, the therapeutic agents may be used in optically pure form.
The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical composition and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable excipient represent a further aspect of the invention.
General Synthetic Routes
The compounds of the invention may be made by a variety of methods. Any previously defined variable will continue to have the previously defined meaning unless otherwise indicated. Illustrative general synthetic methods are set out in the following schemes, and can be readily adapted to prepare other compounds of the invention. Specific compounds of the invention are prepared in the Examples section.
Compounds of formula (I) may be prepared as described in any of the Schemes below:
Figure US10966961-20210406-C00009

wherein R1, R2, R3 and R4 are as described above and X is either Br, Cl or OH.
In respect of the steps shown in Scheme 1 above the following reaction conditions may be utilised:
Step 1: is an amidation of an ester and may be carried out using an appropriate amine of formula R1—NH2, optionally in the presence of a suitable solvent, such as THF, at a suitable temperature, such as 0° C. to r.t.
Step 2: is a Mitsunobu coupling reaction where X=OH and may be carried out using any suitable alcohol, in the presence of a Mitsunobu coupling reagent, such as either DEAD or DIAD with a suitable phosphine, such as Ph3P; or alternately with a suitable phosphorane reagent such as CMBP in a suitable solvent, such as THF or acetonitrile, at a suitable temperature, such as room temperature or at 120° C.
Step 3: is an alkylation reaction where X=Br or Cl and may be carried out using a suitable base, such as potassium carbonate in a suitable solvent, such as acetone, at a suitable temperature, such as room temperature.
Step 4: is a base-mediated ester hydrolysis and may be carried out using any suitable base, such as lithium hydroxide, optionally in a suitable solvent or mixture of solvents, such as 1,4-dioxane and water, at a suitable temperature, such as room temperature.
Step 5: is an amide coupling reaction and may be carried out using an amine reagent, R2—NH2, in the presence of a suitable tertiary amine, such as triethylamine or DIPEA, in the presence of a suitable amide coupling reactant, such as HATU, in a suitable solvent, such as DCM or DMF, at a suitable temperature, such as room temperature.
Step 6: is an acid-mediated ester cleavage and may be carried out using any suitable acid, such as sulfuric acid, optionally in a suitable solvent or mixture of solvents, such as 1,4 dioxane and water, at a suitable temperature, such as heating at reflux.
Step 7: is an amide coupling reaction and may be carried out using an amine reagent, R1—NH2, in the presence of a suitable tertiary amine, such as triethylamine or DIPEA, in the presence of a suitable amide coupling reactant, such as HATU, in a suitable solvent, such as DCM or DMF, at a suitable temperature, such as room temperature.
It will be appreciated by those skilled in the art that it may be advantageous to protect one or more functional groups of the compounds described above. Examples of protecting groups and the means for their removal can be found in T. W. Greene ‘Protective Groups in Organic Synthesis’ (4th edition, J. Wiley and Sons, 2006), incorporated herein by reference as it relates to such procedures.
Suitable amine protecting groups include acyl (e.g. acetyl, carbamate (e.g. 2′,2′,2′-trichloroethoxycarbonyl, benzyloxycarbonyl or t-butoxycarbonyl) and arylalkyl (e.g. benzyl), which may be removed by acid mediated cleavage (e.g. using an acid such as hydrochloric acid in 1,4-dioxane or trifluoroacetic acid in dichloromethane) or reductively (e.g. hydrogenolysis of a benzyl or benzyloxycarbonyl group or reductive removal of a 2′,2′,2′-trichloroethoxycarbonyl group using zinc in acetic acid) as appropriate. Other suitable amine protecting groups include trifluoroacetyl (—C(O)CF3) which may be removed by base catalysed hydrolysis.
It will be appreciated that in any of the routes described above, the precise order of the synthetic steps by which the various groups and moieties are introduced into the molecule may be varied. It will be within the skill of the practitioner in the art to ensure that groups or moieties introduced at one stage of the process will not be affected by subsequent transformations and reactions, and to select the order of synthetic steps accordingly.
Certain intermediate compounds described above form a yet further aspect of the invention.
For any of the hereinbefore described reactions or processes, conventional methods of heating and cooling may be employed, for example temperature-regulated oil-baths or temperature-regulated hot-blocks, and ice/salt baths or dry ice/acetone baths respectively. Conventional methods of isolation, for example extraction from or into aqueous or non-aqueous solvents may be used.
Conventional methods of drying organic solvents, solutions, or extracts, such as shaking with anhydrous magnesium sulfate, or anhydrous sodium sulfate, or passing through a hydrophobic frit, may be employed. Conventional methods of purification, for example crystallisation and chromatography, for example silica chromatography or reverse-phase chromatography, may be used as required. Crystallisation may be performed using conventional solvents such as ethyl acetate, methanol, ethanol, or butanol, or aqueous mixtures thereof. It will be appreciated that specific reaction times and temperatures may typically be determined by reaction-monitoring techniques, for example thin-layer chromatography and LC-MS.
General Experimental Details
All temperatures referred to are in ° C.
As used herein the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. Specifically, the following abbreviations may be used in the examples and throughout the specification:
General Methods
General Experimental Details
As used herein the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. Specifically, the following abbreviations may be used in the examples and throughout the specification:
Abbreviations
  • AcOH acetic acid
  • AMU atomic mass units
  • Aq aqueous
  • BOC/Boc tert-butyloxycarbonyl
  • Cs2CO3 cesium carbonate
  • CHCl3 chloroform
  • CMBP (cyanomethylene)tributylphosphorane
  • CPME cyclopentyl methyl ether
  • CV column volume
  • DCM dichloromethane
  • DIAD diisopropyl azodicarboxylate
  • DIBAL-H diisobutylaluminium hydride
  • DIPEA diisopropylethylamine
  • DMAP 4-dimethylaminopyridine
  • DMF dimethylformamide
  • DMSO dimethylsulfoxide
  • DMSO-d6 deuterated dimethylsulfoxide
  • DPPA diphenylphosphoryl azide
  • Et3N triethylamine
  • EtOAc ethyl acetate
  • EtOH ethanol
  • h hour(s)
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tramethyluronium hexafluorophosphate
  • HBr hydrogen bromide
  • HCl hydrochloric acid
  • HPLC high performance (quid chromatography
  • Isolera Biotage® Flash purification system
  • K2CO3 potassium carbonate
  • LiCl lithium chloride
  • LCMS liquid chromatography-mass spectrometry
  • LiOH lithium hydroxide
  • M molar (concentration)
  • MDAP mass directed autopreparative chromatography
  • MeCN acetonitrile
  • MeOH methanol
  • 2-MeTHF 2-methyltetrahydrofuran
  • minutes(s)
  • MS mass spectrometry
  • Ms-Cl methanesulfonyl chloride
  • N normal (concentration)
  • N2 nitrogen gas
  • NaBH4 sodium borohydride
  • Na2CO3 sodium carbonate
  • NaH sodium hydride
  • NaHCO3 sodium bicarbonate
  • NaOH sodium hydroxide
  • Na2SO4 sodium sulphate
  • NH3 ammonia
  • NH4Cl ammonium chloride
  • NUT nuclear protein in testis
  • obs obscured
  • Ph3P triphenylphosphine
  • RBF round bottomed flask
  • Rt retention time
  • rt room temperature
  • sat saturated
  • SCX Isolute strong cation exchange sorbent SPE
  • SFC supercritical fluid chromatography
  • SiO2 silicon dioxide
  • SNAP Biotage® (silica) flash chromatography cartridge
  • SP4 Biotage® Flash purification system
  • SPE solid phase extraction
  • T3P propylphosphonic anhydride solution
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • TBDMS-Cl tert-butyldimethylsilyl chloride
  • TLC Thin layer chromatography
  • Ts tosyl
  • pTsCl tosyl chloride
  • UPLC ultra performance liquid chromatograpy
  • UV ultraviolet
  • XantPhos 1,1′-(9,9-dimethyl-9H-xanthene-4,5-diyl)bis[1,1-diphenylphosphine
The names of the following compounds have been obtained using the compound naming programme “ACD Name Pro 6.02” or using the naming functionality of ChemDraw Ultra 12.0.
LCMS Methodology
Formic Method
LC Conditions
The UPLC analysis was conducted on an Acquity UPLC CSH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 40° C.
The solvents employed were:
A=0.1% v/v solution of formic acid in water
B=0.1% v/v solution of formic acid in acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 1 97 3
1.5 1 5 95
1.9 1 5 95
2.0 1 97 3
The UV detection was a summed signal from wavelength of 210 nm to 350 nm.
MS conditions
MS Waters ZQ
Ionisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.27 sec
Inter scan delay 0.10 sec
High pH Method
LC Conditions
The UPLC analysis was conducted on an Acquity UPLC CSH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 40° C.
The solvents employed were:
A=10 mM ammonium hydrogen carbonate in water adjusted to pH10 with ammonia solution
B=acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 1 97 3
0.05 1 97 3
1.5 1 5 95
1.9 1 5 95
2.0 1 97 3
The UV detection was a summed signal from wavelength of 210 nm to 350 nm.
MS Conditions
MS Waters ZQ
Ionisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.27 sec
Inter scan delay 0.10 sec
TFA Method
LC conditions
The UPLC analysis was conducted on an Acquity UPLC CSH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 40° C.
The solvents employed were:
A=0.1% v/v solution of trifluoroacetic acid in water
B=0.1% v/v solution of trifluoroacetic acid in acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 1 95 5
1.5 1 5 95
1.9 1 5 95
2.0 1 95 5
The UV detection was a summed signal from wavelength of 210 nm to 350 nm.
MS conditions
MS Waters ZQ
Ionisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.27 sec
Inter scan delay 0.10 sec
Method A
LC conditions
The UPLC analysis was conducted on an Acquity BEH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 35° C.
The solvents employed were:
A=0.1% v/v solution of formic acid in water
B=0.1% v/v solution of formic acid in acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 0.6 97 3
0.4 0.6 97 3
3.2 0.6 2 98
3.8 0.6 2 98
4.2 0.6 97 3
4.5 0.6 97 3

Method B
LC Conditions
The UPLC analysis was conducted on an Acquity BEH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 35° C.
The solvents employed were:
A=0.05% v/v solution of formic acid in water
B=0.05% v/v solution of formic acid in acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 0.6 97 3
0.4 0.6 97 3
3.2 0.6 2 98
3.8 0.6 2 98
4.2 0.6 97 3
4.5 0.6 97 3

Method C
LC Conditions
The UPLC analysis was conducted on a Xbridge C18 column (150 mm×4.6 mm, i.d. 3.5 μm packing diameter) at 35° C.
The solvents employed were:
A=0.05% v/v solution of trifluoroacetic acid in water
B=acetonitrile
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 1.0 95 5
0.5 1.0 95 5
7 1.0 5 95
14 1.0 5 95
14.5 1.0 95 5
15 1.0 95 5

Method D
LC Conditions
The UPLC analysis was conducted on an Acquity BEH C18 column (100 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 35° C.
The solvents employed were:
A=0.05% v/v solution of formic acid in acetonitrile
B=0.05% v/v solution of formic acid in water
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 0.45 3 97
0.4 0.45 3 97
7.5 0.45 98 2
9.5 0.45 98 2
9.6 0.45 3 97
10 0.45 3 97

Method E
LC Conditions
The UPLC analysis was conducted on an Acquity BEH C18 column (50 mm×2.1 mm, i.d. 1.7 μm packing diameter) at 35° C.
The solvents employed were:
A=0.05% v/v solution of formic acid in acetonitrile
B=0.05% v/v solution of formic acid in water
The gradient employed was:
Time (min) Flow rate (mL/min) % A % B
0 0.45 3 97
0.4 0.45 3 97
4.0 0.45 98 2
4.5 0.45 97.5 2.5
5.0 0.45 3 97
5.5 0.45 3 97

General MDAP Purification Methods
Listed below are examples of mass-directed autopreparative chromatography (MDAP) methods that have been used or may be used in compound purification.
MDAP (High pH). The HPLC analysis was conducted on an Xselect CSH C18 column (150 mm×30 mm i.d. 5 μm packing diameter) at ambient temperature, eluting with 10 mM ammonium bicarbonate in water adjusted to pH 10 with ammonia solution (Solvent A) and acetonitrile (Solvent B) using an elution gradient of between 0 and 100% Solvent B over 15 or 25 min.
The UV detection was an averaged signal from wavelength of 210 nm to 350 nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
MDAP (Formic). The HPLC analysis was conducted on an Xselect CSH C18 column (150 mm×30 mm i.d. 5 μm packing diameter) at ambient temperature, eluting with 0.1% formic acid in water (Solvent A) and 0.1% formic acid in acetonitrile (Solvent B) using an elution gradient of between 0 and 100% solvent B over 15 or 25 min.
The UV detection was an averaged signal from wavelength of 210 nm to 350 nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
MDAP (TFA). The HPLC analysis was conducted on an Xselect CSH C18 column (150 mm×30 mm i.d. 5 μm packing diameter) at ambient temperature, eluting with 0.1% v/v solution of trifluoroacetic acid in water (Solvent A) and 0.1% v/v solution of trifluoroacetic acid in acetonitrile (Solvent B) using an elution gradient of between 0 and 100% solvent B over 15 or 25 min.
The UV detection was an averaged signal from wavelength of 210 nm to 350 nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using alternate-scan positive and negative electrospray. Ionisation data was rounded to the nearest integer.
NMR
Spectra were run on either a 400 MHz or 600 MHz NMR machine at 302 K.
Intermediate 1: Methyl 3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00010
A solution of dimethyl 1H-pyrazole-3,5-dicarboxylate (10 g, 54.3 mmol, commercially available from, for example, Fluorochem) in methanamine (5% wt solution in water) (67.5 g, 109 mmol, commercially available from, for example, Spectrochem) was stirred under nitrogen at 0° C. The reaction mixture was stirred at 0° C. for 30 min. After 16 h the reaction mixture was neutralized with 1N HCl, extracted with EtOAc (500 mL) and washed with brine (100 mL). The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated to afford methyl 5-(methylcarbamoyl)-1H-pyrazole-3-carboxylate (4 g, 14.19 mmol, 26% yield) of the crude compound. The reaction was repeated again on the same scale to afford a further 3.8 g of crude compound and again using a solution of dimethyl 1H-pyrazole-3,5-dicarboxylate (20 g, 54.3 mmol) in methanamine (5% wt solution in water) (67.5 g, 109 mmol) stirred under nitrogen at 0° C. using the same work-up as above to afford a further 7 g of crude compound. The combined crude batches were purified by column chromatography (100-200 silica gel) eluting with 0-10% MeOH in DCM. The desired fractions were collected and dried under reduced pressure to afford the crude compound (10 g). This was diluted with anhydrous methanol (100 mL) and kept at rt for 24 h. After 24 h the solid was filtered through a Buchner funnel, washed with cooled anhydrous methanol (20 mL) and the residue was then washed with pentane and dried to afford methyl 5-(methylcarbamoyl)-1H-pyrazole-3-carboxylate (8 g, 43.6 mmol, 20% yield) as a off white solid.
1H NMR (400 MHz, DMSO-d6) δ ppm 14.27 (br. s., 1H) 8.42 (br. s., 1H) 7.22 (br. s., 1H) 3.83 (s, 3H) 2.76 (d, J=4.6 Hz, 3H)
LCMS (2 min Formic): Rt=0.44 min, [MH]+=184.1.
Intermediate 2: Methyl (S)-dimethyl 1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxylate
Figure US10966961-20210406-C00011
Diisopropyl (E)-diazene-1,2-dicarboxylate (8.24 g, 40.7 mmol) was added dropwise to a suspension of dimethyl 1H-pyrazole-3,5-dicarboxylate (5 g, 27.2 mmol, commercially available from, for example, Fluorochem), (R)-1-phenylethan-1-ol (3.98 g, 32.6 mmol, commercially available from, for example, Sigma Aldrich) and triphenylphosphine polymer bound 3 mmol/g (13.70 g, 40.7 mmol, commercially available from, for example, Sigma Aldrich) in 2-MeTHF (50 mL) at 0° C. under nitrogen. The mixture was stirred overnight, allowing it to warm to rt. In a separate reaction, dimethyl 1H-pyrazole-3,5-dicarboxylate (2 g, 10.86 mmol), (R)-1-phenylethan-1-ol (1.592 g, 13.03 mmol) and triphenylphosphine polymer bound 3 mmol/g (5.48 g, 16.29 mmol) were combined in a RBF, 2-MeTHF (50 mL) was added and the mixture was stirred under nitrogen, cooling in an ice bath for 30 min, then diisopropyl (E)-diazene-1,2-dicarboxylate (3.29 g, 16.29 mmol) was added dropwise over 30 min and the resulting mixture stirred overnight, allowing it to warm to rt. The reaction mixtures were combined and the combined suspension was filtered and the solid polymer bound triphenylphosphine(oxide) was washed with EtOAc (100 mL). Then the combined organics were washed with water (200 mL), dried and evaporated in vacuo and the residue purified by chromatography on a 340 g silica column eluting with 0-25% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give dimethyl (S)-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxylate (7.2 g, 24.97 mmol, 66% yield) as a colourless oil.
1H NMR (400 MHz, CHLOROFORM-d) δ ppm 7.39 (s, 1H) 7.29-7.33 (m, 4H) 7.22-7.28 (m, 1H) 6.63 (q, J=7.1 Hz, 1H) 3.96 (s, 3H) 3.86 (s, 3H) 1.99 (d, J=7.1 Hz, 3H) LCMS (2 min High pH): Rt=1.13 min, [MH]+=289.2.
Intermediate 3: (S)-5-(Methoxycarbonyl)-1-(1-phenylethyl)-1H-pyrazole-3-carboxylic acid
Figure US10966961-20210406-C00012
Dimethyl (S)-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxylate (15 g, 52.0 mmol) was dissolved in 1,4-dioxane (60 mL), then water (120 mL) was added followed by dropwise addition of H2SO4 (1.664 mL, 31.2 mmol). The mixture was heated at reflux for 3 days, then cooled to rt and extracted with EtOAc (2×100 mL). The combined organics were dried and evaporated in vacuo to give a colourless gum, which was dissolved in DCM and loaded onto a 340 g silica column, then eluted with 0-50% (1% AcOH/EtOAc)/cyclohexane and the product-containing fractions were evaporated in vacuo to give (S)-5-(methoxycarbonyl)-1-(1-phenylethyl)-1H-pyrazole-3-carboxylic acid (6.7 g, 24.43 mmol, 47% yield) as a colourless solid.
1H NMR (400 MHz, CHLOROFORM-d) δ ppm 7.46 (s, 1H) 7.24-7.37 (m, 5H) 6.66 (q, J=7.1 Hz, 1H) 3.88 (s, 3H) 2.00 (d, J=7.1 Hz, 3H). 1 exchangeable proton not observed.
LCMS (2 min Formic): Rt=0.96 min, [MH]+=275.3.
Intermediate 4: (S)-Methyl 3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00013
(S)-5-(Methoxycarbonyl)-1-(1-phenylethyl)-1H-pyrazole-3-carboxylic acid (2 g, 7.29 mmol) was dissolved in DCM (20 mL) and Et3N (1.525 mL, 10.94 mmol) and HATU (3.33 g, 8.75 mmol) were added, followed by methanamine (2M in THF) (3.65 mL, 7.29 mmol) and the mixture was stirred for 2 h at rt, then washed with water (20 mL) and 0.5M HCl (20 mL). The organic layer was dried and evaporated in vacuo and the residue purified by chromatography on a 50 g silica column to give methyl (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylate (2.1 g, 7.31 mmol, 100% yield) as a colourless gum.
1H NMR (400 MHz, CHLOROFORM-d) δ ppm 7.38 (s, 1H) 7.23-7.35 (m, 5H) 6.96 (d, J=3.7 Hz, 1H) 6.63 (q, J=7.1 Hz, 1H) 3.86 (s, 3H) 3.03 (s, 3H) 1.92 (d, J=7.1 Hz, 3H)
LCMS (2 min Formic): Rt=0.99 min, [MH]+=288.2.
Intermediate 5: (S)-3-(Methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00014
Methyl (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylate (4.2 g, 14.62 mmol) was dissolved in a mixture of methanol (10 mL) and THF (10 mL), then NaOH (2M aqueous solution, 14.62 mL, 29.2 mmol) was added and the mixture was stirred for 3 h at rt. The solvent was evaporated in vacuo, the residue dissolved in water (30 mL) and washed with ether, then the aqueous layer acidified with 2M HCl to pH 4 and the resulting mixture stirred for 20 min while cooling in an ice bath. The resulting solid was collected by filtration to give (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1/pyrazole-5-carboxylic acid (3.3 g, 12.07 mmol, 83% yield) as a colourless solid.
1H NMR (400 MHz, DMSO-d6) δ ppm 13.62 (br. s., 1H) 8.26 (d, J=4.6 Hz, 1H) 7.18-7.37 (m, 5H) 7.14 (s, 1H) 6.61 (q, J=6.8 Hz, 1H) 2.77 (d, J=4.6 Hz, 3H) 1.80-1.91 (m, 3H) LCMS (2 min Formic): Rt=0.86 min, [MH]+=274.2.
Intermediate 6: Dimethyl 1-benzyl-1H-pyrazole-3,5-dicarboxylate
Figure US10966961-20210406-C00015
To a solution of dimethyl 1H-pyrazole-3,5-dicarboxylate (10 g, 54.3 mmol) in acetone (100 mL) stirred under nitrogen at 0° C. was added K2CO3 (15.01 g, 109 mmol), followed by the addition of benzyl bromide (7.10 mL, 59.7 mmol) dropwise over 1 min. The reaction mixture was stirred at rt for 5 h. The reaction was filtered and the filtrate was concentrated under vaccum to afford the crude product. The crude product was triturated with n-pentane (3×10 mL), then dried under vaccum to afford pure dimethyl 1-benzyl-1H-pyrazole-3,5-dicarboxylate (8.5 g, 31.0 mmol, 57% yield) as a white solid.
LCMS (10 min Method D): Rt=4.87 min, [MH]+=275.0.
Intermediate 7: 1-Benzyl-5-(methoxycarbonyl)-1H-pyrazole-3-carboxylic acid
Figure US10966961-20210406-C00016
To a solution of dimethyl 1-benzyl-1H-pyrazole-3,5-dicarboxylate (30 g, 109 mmol) in 1,4-dioxane (140 mL) and water (280 mL) stirred under nitrogen at rt was added conc. H2SO4 (2.92 mL, 54.7 mmol, 18.4M) in one charge. The reaction mixture was stirred at reflux for 72 h. The reaction was then cooled to Rt and extracted with EtOAc (3×125 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to afford the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 15% EtOAc in n-hexane and collected starting material fractions were concentrated under reduced pressure to get recovered starting material (15 g) as an off-white solid. The column was then eluted with 60% EtOAc in hexane and collected pure fractions were concentrated under vacuum to provide pure 1-benzyl-5-(methoxycarbonyl)-1H-pyrazole-3-carboxylic acid (6 g) as a white solid. Impure fractions were also isolated to give a further 10 g of crude mixture which was added to a silica gel 60-120 column and was eluted with 60% EtOAc in hexane to provide further pure fractions. These were concentrated under vacuum to get a second batch of pure required product (5.5 g) which was combined to give 1-benzyl-5-(methoxycarbonyl)-1H-pyrazole-3-carboxylic acid (11.5 g, 44.0 mmol, 40% yield) as an off-white solid. Then the column was flushed with EtoAc to give a further batch of impure product (3.4 g).
LCMS (10 min Method D): Rt=4.89 min, [MH]+=261.0.
Intermediate 8: Methyl 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00017
To a solution of 1-benzyl-5-(methoxycarbonyl)-1H-pyrazole-3-carboxylic acid (13.6 g, 50.7 mmol) and DIPEA (26.6 mL, 152 mmol) in DMF (100 mL) stirred under nitrogen at rt was added HATU (28.9 g, 76 mmol), followed by the addition of methanamine hydrochloride (4.11 g, 60.8 mmol) in one charge over 1 min. The reaction mixture was stirred at rt for 16 h. The reaction was poured into ice water, then extracted with EtOAc (3×100 mL). The combined organic layer was washed with water (3×50 mL), brine solution, then dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to afford the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 65% EtOAc in n-hexane and the collected pure fractions were concentrated under reduced pressure to get methyl 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (12.1 g, 42.7 mmol, 84% yield) as an off-white solid.
LCMS (5.5 min Method E): Rt=2.32 min, [MH]+=274.1.
Intermediate 9: 1-Benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00018
To a solution of methyl 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (12.1 g, 42.7 mmol) in THF (70 mL) and water (70 mL) stirred under nitrogen at rt was added LiOH (5.11 g, 213 mmol) in one charge over 1 min. The reaction mixture was stirred at rt for 2 h. The reaction was then diluted with water (20 mL), then the aqueous layer was washed with EtOAc (3×15 mL). Then the aqueous layer pH was adjusted with 2NHCl to pH 1, then this was extracted with EtoAc (3×100 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to afford the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 85% EtOAc in n-hexane and collected pure fractions were concentrated under reduced pressure to afford the desired product, 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (8.5 g, 32.8 mmol, 77% yield) as an off-white solid.
LCMS (10 min Method D): Rt=3.25 min, [MH]+=260.1.
Intermediate 10: N3-Cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00019
To a solution of 1-benzyl-N5-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide (500 mg, 1.525 mmol, for a preparation, see example 92) in ethanol (1 mL) stirred under nitrogen at rt was added palladium hydroxide on carbon (100 mg, 0.712 mmol) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction was filtered through the Celite bed, then washed with ethanol (25 mL) and the filtrate was concentrated under vacuum to afford the crude product. The crude product was triturated with diethyl ether (3×1 mL), to afford crystalline solid, filtered and the solid was dried under vacuum to get N5-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide (250 mg, 1.045 mmol, 68.5% yield) as a white solid.
LCMS (5.5 min Method E): Rt=1.29 min, [MH]+=209.3.
Intermediate 11: N5-Cyclopropyl-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00020
To a solution of N5-cyclopropyl-N3-methylpyrazole-3,5-dicarboxamide (250 mg, 1.045 mmol) in DMF (2 mL) stirred under nitrogen at rt was added K2CO3 (433 mg, 3.13 mmol) followed by the addition of (1-bromoethyl)benzene (232 mg, 1.253 mmol) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction mass was poured into ice water, then extracted with EtOAc (3×20 mL). The combined organic layer was washed with water (3×15 mL), brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to afford the crude product. This showed two regioisomers.
Regio-isomer 1
LCMS (10 min Method D): Rt=3.40 min, [MH]+=313.1.
Regio-isomer 2
LCMS (10 min Method D): Rt=3.48 min, [MH]+=313.1.
Intermediate 12: Methyl 1-tosyl-1H-indole-5-carboxylate
Figure US10966961-20210406-C00021
To a solution of methyl 1H-indole-5-carboxylate (2 g, 11.42 mmol) in DMF (15 mL) stirred under nitrogen at 0° C. was added NaH (0.548 g, 13.70 mmol, 60% dispersion in mineral oil) portion wise, then the reaction mixture was stirred for 10 min at the same temperature, then at rt for 30 min, after which was added pTsCl (2.61 g, 13.70 mmol) portionwise over 1 min. The reaction mixture was stirred at rt for 2 h. The reaction was poured into ice water, then extracted with EtOAc (3×25 mL). The combined organic layer was washed with cold water (3×15 mL), brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to afford the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 15% EtOAc in n-hexane and the collected pure fractions were concentrated under reduced pressure to afford methyl 1-tosyl-1H-indole-5-carboxylate (1.9 g, 5.59 mmol, 49.0% yield) as an off-white solid.
LCMS (5.5 min Method E): Rt=3.27 min, [MH]+=330.0.
Intermediate 13: (1-Tosyl-1H-indol-5-yl)methanol
Figure US10966961-20210406-C00022
To a solution of methyl 1-tosyl-1H-indole-5-carboxylate (1.9 g, 5.59 mmol) in DCM (20 mL) stirred under nitrogen at rt was added a solution of DIBAL-H (25.2 mL, 25.2 mmol, 1M solution in Toluene) dropwise over 1 min. The reaction mixture was stirred at −78° C. for 2.5 h. The reaction was then quenched with methanol (0.226 mL, 5.59 mmol) at −78° C. and then allowed to warm up to ambient temperature. The reaction mass was diluted with saturated Rochelle's salt solution (120 mL) and stirred for 16 h, then the layers were separated, and the aqueous phase was extracted with DCM (2×100 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to afford the crude product. The crude product was triturated with n-pentane (3×2 mL) then dried under vaccum to afford (1-tosyl-1H-indol-5-yl)methanol (1.70 g, 5.41 mmol, 97% yield) as an off-white solid.
LCMS (5.5 min Method E): Rt=2.72 min, [MH]+=300.0.
Intermediate 14: 5-(Bromomethyl)-1-tosyl-1H-indole
Figure US10966961-20210406-C00023
To a solution of (1-tosyl-1H-indol-5-yl)methanol (1.70 g, 5.41 mmol) in DCM (20 mL) stirred under nitrogen at rt was added a solution of HBr (0.294 mL, 5.41 mmol) dropwise during 1 min. The reaction mixture was stirred at rt for 4 h. The reaction was poured into ice water, then stirred for 10 min at rt, then filtered and the solid product was dried under vacuum to give 5-(bromomethyl)-1-tosyl-1H-indole (1.48 g, 3.49 mmol, 65% yield) as an off-white solid.
LCMS (5.5 min Method E): Rt=3.47 min, [MH]+=365.9.
Intermediate 15: N5-Cyclopropyl-N3-methyl-1-((1-tosyl-1H-indol-5-yl)methyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00024
To a solution of N5-cyclopropyl-N3-methylpyrazole-3,5-dicarboxamide (300 mg, 1.366 mmol) in DMF (2 mL) stirred under nitrogen at rt was added K2CO3 (566 mg, 4.10 mmol), followed by the addition of 5-(bromomethyl)-1-tosyl-1H-indole (703 mg, 1.640 mmol) dropwise over 1 min. The reaction mixture was stirred at rt for 1 h. The reaction was poured into ice water, then extracted with EtOAc (3×15 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to give the crude product (420 mg) as a mixture of regioisomers which were used in the next step without purification.
LCMS (5.5 min Method E): Rt=2.82 min, [MH]+=492.0. Regioisomer 1
LCMS (5.5 min Method E): Rt=2.85 min, [MH]+=492.0. Regioisomer 2
Intermediate 16: N5-Cyclopropyl-N3-methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00025
To a solution of N5-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide (300 mg, 1.366 mmol) in acetone (1 mL) stirred under nitrogen at rt was added K2CO3 (566 mg, 4.10 mmol) followed by the addition of 4-(bromomethyl)-1-tosyl-1H-indole (597 mg, 1.640 mmol) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction was poured into ice water, then extracted with EtOAc (3×25 mL). The combined organic layers were washed with water (3×15 mL), brine solution, dried over sodium sulpate, filtered and the filtrate was concentrated under vaccum to give the crude product as a mixture of regioisomers (550 mg) which were used in the next step without purification.
LCMS (10 min Method D): Rt=4.39 min, [MH]+=492.1. Regioisomer 1
LCMS (10 min Method D): Rt=4.44 min, [MH]+=492.1. Regioisomer 2
Intermediate 17: N3,N5-Dimethyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00026
1H-Pyrazole-3,5-dicarboxylic acid (200 mg, 1.281 mmol), methanamine (2M in THF, 0.705 mL, 1.409 mmol) and HATU (536 mg, 1.409 mmol) were combined in DMF (2 mL). DIPEA (0.448 mL, 2.56 mmol) was added and reaction mixture stirred at rt. under N2 for 2 h. The reaction mixture was concentrated to dryness and used crude for the next step.
LCMS (2 min Formic): Rt=0.37 min, [MH]+=183.1.
Intermediate 18: 4-Bromo-1-tosyl-1H-pyrrolo[2,3-c]pyridine
Figure US10966961-20210406-C00027
4-Bromo-1H-pyrrolo[2,3-c]pyridine (5 g, 25.4 mmol, commercially available from, for example, Fluorochem) was dissolved in DMF (50 mL) and cooled in an ice bath under nitrogen, then NaH (a 60% suspension in mineral oil, 1.319 g, 33.0 mmol) was added in small portions and the mixture was stirred for 20 min before addition of tosyl chloride (5.32 g, 27.9 mmol). The mixture was stirred overnight, allowing it to warm to rt, then the mixture was diluted with water (100 mL) and stirred for 30 min, giving a white suspension. This was filtered and the solid washed with water, then dried to give a colourless solid. The crude product was suspended in ether (20 mL) and stirred for 5 min, then diluted with cyclohexane (20 mL) and filtered to give the desired product (7.9 g, 22.49 mmol, 89% yield) as a colourless solid.
LCMS (2 min High pH): Rt=1.24 min, [MH]+=351/353
Intermediate 19: Methyl 1-tosyl-1H-pyrrolo[2,3-c]pyridine-4-carboxylate
Figure US10966961-20210406-C00028
4-Bromo-1-tosyl-1H-pyrrolo[2,3-c]pyridine (7.8 g, 22.21 mmol) palladium(II) acetate (0.499 g, 2.221 mmol) and xantphos (1.285 g, 2.221 mmol) were combined in a round-bottomed flask, which was sealed with a suba seal and purged with nitrogen. DMF (20 mL), Et3N (9.29 mL, 66.6 mmol) and methanol (17.97 mL, 444 mmol) were added, the vessel was purged with carbon monoxide from a balloon, then heated under a CO atmosphere overnight. The mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL), the organic layer washed with 10% aq LiCl solution, dried and evaporated in vacuo and the residue purified by chromatography on a 50 g silica column eluting with 0-100% EtOAc/cyclohexane to give the desired product (4.4 g, 13.32 mmol, 60% yield)
LCMS (2 min High pH): Rt=1.12 min, [MH]+=331.1
Intermediate 20: (1-Tosyl-1H-pyrrolo[2,3-c]pyridin-4-yl)methanol
Figure US10966961-20210406-C00029
Methyl 1-tosyl-1H-pyrrolo[2,3-c]pyridine-4-carboxylate (510 mg, 1.544 mmol) was taken up in DCM (15 mL) under nitrogen and cooled to −78° C. DIBAL-H (1M in THF, 15.44 mL, 15.44 mmol) was added to the reaction dropwise and it was left to stir at −78° C. for 3 h. The reaction was quenched with methanol (15 mL added dropwise) at −78° C. and then allowed to warm to rt. Rochelle's salt (saturated in 25 mL of water) was added and the mixture was stirred for 45 min. The mixture was separated, and the organic layer was washed with brine (50 mL), filtered through a hydrophobic frit and concentrated in vacuo to yield a yellow solid. This solid was taken up in DCM (15 mL), put under nitrogen and cooled to −78° C. DIBAL-H (1M in THF, 15.44 mL, 15.44 mmol) was added dropwise, and then the reaction was allowed to warm up to −40° C. It was left to stir at this temperature for 5 h. The reaction was quenched with methanol (15 mL added dropwise) at −40° C. and then allowed to warm to rt. Rochelle's salt (saturated in 30 mL of water) was added and the mixture was stirred for 45 min. The mixture was separated, and the organic layer was washed with brine (2×25 mL), filtered through a hydrophobic frit and concentrated in vacuo to yield a yellow oil (486 mg). The crude product was purified by silica gel column chromatography, eluting with 5-50% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield the desired product (148 mg, 0.416 mmol, 27% yield) as a white solid
LCMS (2 min Formic): Rt=0.57 min, [MH]+=303.1
Intermediate 21: 1-(1-Tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)ethanone
Figure US10966961-20210406-C00030
1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-ethan-1-one (500 mg, 3.12 mmol, commercially available from, for example, Activate Scientific) was dissolved in DMF (5 mL) and cooled to 0° C. in an ice-bath under a nitrogen atmosphere. Sodium hydride (60% in mineral oil, 150 mg, 3.75 mmol) was added portionwise, and the reaction mixture was stirred at 0° C. for 30 min before warming to rt for 30 min. Tosyl chloride (714 mg, 3.75 mmol) was added giving an orange suspension (exothermic addition) and the reaction mixture stirred at rt for 15 min. The reaction mixture was cooled to 0° C. in an ice-bath and quenched with the dropwise addition of water (3 mL). This was then partitioned between ethyl acetate (20 mL) and sat. aq. LiCl solution (20 mL). The organic layer was separated and the aq. layer further extracted with ethyl acetate. The combined organic layers were dried (Na2SO4) and conc. to give ˜1.5 g crude brown residue. This was purified by silica gel column chromatography eluting with 5-50% ethyl acetate/cyclohexane to give the desired product (438 mg, 1.254 mmol, 40% yield) as a pale yellow solid.
LCMS (2 min Formic): Rt=1.12 min, [MH]+=315
Intermediate 22: 1-(1-Tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-ethanol
Figure US10966961-20210406-C00031
To a suspension of 1-(1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-ethan-1-one (438 mg, 1.393 mmol) in ethanol (10 mL) was added THF (4 mL) to solubilise the reaction mixture, followed by sodium borohydride (58.0 mg, 1.533 mmol) in an ice-bath. The reaction mixture was stirred under a nitrogen atmosphere at rt for 1 h. 1M HCl solution (20 mL) was added and the aqueous layer extracted with DCM. The combined organic layers were dried (hydrophobic frit) and concentrated to give the desired product (493 mg, 1.325 mmol, 95% yield) as a pale yellow oil.
LCMS (2 min Formic): Rt=0.98 min, [MH]+=317
Intermediate 23: Methyl 1-tosyl-1H-indole-5-carboxylate
Figure US10966961-20210406-C00032
Methyl 1H-indole-5-carboxylate (4.31 g, 24.60 mmol, commercially available from, for example, Sigma Aldrich) was taken up in DMF (50 mL) and cooled to 0° C. NaH (60% suspension in mineral oil, 1.181 g, 29.5 mmol) was added in small portions, and the reaction was left to stir for 1 h. Tosyl chloride (5.63 g, 29.5 mmol) was added, and the reaction was allowed to warm to rt and stirred overnight. Additional NaH (0.480 g, 60% suspension in mineral oil) was added, and the reaction left to stir at rt for 1 h. The reaction was quenched with water (25 mL). Water (175 mL) was added, and the reaction was extracted with ethyl acetate (2×250 mL). The combined organics were washed with brine (200 mL), dried with Na2SO4, filtered and concentrated in vacuo to yield a brown solid. 1:1 MeOH:DCM (50 mL) was added to the crude product, and free flow silica added (5 g). The solvent was removed in vacuo and the silica loaded onto a 100 g silica gel column cartridge and eluted with 5-25% ethyl acetate in cyclohexane. The fractions were combined and concentrated in vacuo. The crude product was applied to a 340 g silica gel column cartridge in the minimum of DCM and eluted with 0-25% ethyl acetate in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield the desired product (6.463 g, 18.64 mmol, 76% yield) as a white solid.
LCMS (2 min Formic): Rt=1.27 min, [MH]+=330.4
Intermediate 24: 1-(1-Tosyl-1H-indol-5-yl)ethanone
Figure US10966961-20210406-C00033
1-(1H-Indol-5-yl)-ethan-1-one (1 g, 6.28 mmol, commercially available from, for example, Fluorochem) was taken up in DMF (10 mL) and cooled to 0° C. NaH (0.302 g, 7.54 mmol, 60% dispersion in mineral oil) was added in small portions and allowed to stir for 15 min. tosyl-Cl (1.437 g, 7.54 mmol) was added, and the reaction was allowed to warm to rt and left to stir for 1.5 h. Additional NaH (63 mg, 0.25 eq., 60% dispersion in mineral oil) was added, and the reaction was left to stir for 30 min. The reaction was quenched with water (40 mL). The reaction mixture was extracted with ethyl acetate (2×50 mL) and the combined organics were washed with brine (75 mL), dried with Na2SO4, filtered and concentrated in vacuo to yield a brown solid. The crude product was applied to a 100 g ULTRA SNAP cartridge in the minimum of DCM and eluted with 0-5% ethyl acetate in DCM. The appropriate fractions were combined and concentrated in vacuo to yield 1-(1-tosyl-1H-indol-5-yl)-ethan-1-one (1.408 g, 4.27 mmol, 68% yield) as a cream solid.
LCMS (2 min High pH): Rt=1.20 min, [MH]+=314.3.
Intermediate 25: 1-(1-Tosyl-1H-indol-5-yl)-ethanol
Figure US10966961-20210406-C00034
1-(1-Tosyl-1H-indol-5-yl)-ethan-1-one (1.394 g, 4.45 mmol) was taken up in THF (40 mL), cooled to 0° C. in an ice bath, and put under nitrogen. sodium borohydride (0.337 g, 8.90 mmol) was added, and the reaction left to stir at rt overnight. Additional sodium borohydride (0.169 g) was added, and the reaction left to stir for 2.5 h. The reaction was cooled to 0° C. and slowly quenched with 1N HCl (20 mL). It was then extracted with DCM (2×25 mL), and the combined organics were filtered through a hydrophobic frit and concentrated in vacuo to yield 1-(1-tosyl-1H-indol-5-yl)-ethan-1-ol (1.398 g, 4.21 mmol, 95% yield) as a colourless gum.
LCMS (2 min High pH): Rt=1.11 min, [M−H]=314.3.
Intermediate 26: (1-Tosyl-1H-indol-5-yl)methanol
Figure US10966961-20210406-C00035
Methyl 1-tosyl-1H-indole-5-carboxylate (1.027 g, 3.12 mmol) was taken up in THF (25 mL) under nitrogen, and cooled to −78° C. DIBAL-H (1M in THF, 8 mL, 8.00 mmol) was added dropwise over 10 min and the reaction was left to stir at −78° C. for 4 h. The reaction was allowed to warm to rt and was left to stir overnight. The reaction was cooled to −78° C. and DIBAL-H (1M in THF, 6.5 mL, 6.50 mmol) was added dropwise over 10 min. The reaction was then allowed to warm to rt and was left to stir for 1.5 h. The reaction was cooled to 0° C. and quenched with MeOH (10 mL added dropwise). Saturated Rochelle's salts (50 mL) was added, and the reaction allowed to warm to rt. Water (50 mL) and DCM (50 mL) were added to the reaction mixture, and the layers were separated. The organic layer was washed with brine (50 mL), eluted through a hydrophobic frit and concentrated in vacuo to yield a colourless oil. The crude product was applied to a silica gel column in the minimum of DCM and eluted with 5-50% ethyl acetate in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield the desired product (876 mg, 2.76 mmol, 89% yield) as a colourless gum.
LCMS (2 min Formic): Rt=0.98 min, [MH]+=317
Intermediate 27: 1-(1-Tosyl-1H-indol-4-yl)ethanone
Figure US10966961-20210406-C00036
1-(1H-Indol-4-yl)-ethan-1-one (500 mg, 3.14 mmol, commercially available from, for example, Activate Scientific) was dissolved in DMF (5 mL) and cooled to 0° C. in an ice-bath under a nitrogen atmosphere. Sodium hydride (151 mg, 3.77 mmol, 60% dispersion in mineral oil) was added portionwise, and the reaction mixture was stirred at 0° C. for 30 min before warming to rt for 30 min. Tosyl chloride (719 mg, 3.77 mmol) was added giving an orange suspension (exothermic addition) and reaction mixture stirred at rt for 15 min. The reaction mixture was cooled to 0° C. in an ice-bath and quenched by the dropwise addition of water (3 mL). This was then partitioned between ethyl acetate (20 mL) and sat. aq. LiCl solution (20 mL). The organic layer was separated and aq. layer further extracted with ethyl acetate. The combined organic layers were dried (Na2SO4) and conc. to give 1.11 g of crude brown residue. This was purified by chromatography on silica gel, eluting with 5-30% ethyl acetate/cyclohexane to give the desired product (809 mg, 2.323 mmol, 74% yield) as a white solid.
LCMS (2 min Formic): Rt=1.24 min, [MH]+=314
Intermediate 28: 1-(1-Tosyl-1H-indol-4-1)ethanol
Figure US10966961-20210406-C00037
To a suspension of 1-(1-tosyl-1H-indol-4-yl)-ethan-1-one (803 mg, 2.56 mmol) in ethanol (20 mL) was added sodium borohydride (107 mg, 2.82 mmol) in an ice-bath. The reaction mixture was stirred under a nitrogen atmosphere at rt for 1.5 h. THF (17 mL) was added to solubilise the reaction mixture which was stirred at rt for a further 2 h. A further portion of sodium borohydride (107 mg, 2.82 mmol) was added and reaction mixture stirred at rt overnight. 1M HCl solution (20 mL) was added and the aqueous layer extracted with DCM. The combined organic layers were dried (hydrophobic frit) and concentrated to give the desired product (797 mg, 2.274 mmol, 89% yield) as a pink solid.
LCMS (2 min Formic): Rt=1.12 min, [MH]=314.2
Intermediate 29: Methyl 1-tosyl-1H-indole-4-carboxylate
Figure US10966961-20210406-C00038
Methyl 1H-indole-4-carboxylate (5 g, 28.5 mmol, commercially available from, for example, Alfa Aesar) was taken up in DMF (50 mL) and cooled in an ice-bath. NaH (60% suspension in mineral oil, 1.370 g, 34.2 mmol) was added portionwise and the reaction left to stir for 30 min. Tosyl chloride (6.53 g, 34.2 mmol) was added and the reaction left to stir and warm up overnight. The reaction was cooled in an ice-bath and carefully quenched with water (200 mL). The mixture was extracted with EtOAc (2×250 mL). The combined organics were washed with brine (200 mL) and eluted through a hydrophobic frit then concentrated in vacuo to give a brown solid. The crude product was applied to a 100 g silica gel cartridge in the minimum of DCM and eluted with 5-25% ethyl acetate in cyclohexane. The appropriate fractions were concentrated in vacuo to give the desired product (6.554 g, 18.90 mmol, 66% yield) as a white solid.
LCMS (2 min High pH): Rt=1.32 min, [MH]+=330.2
Intermediate 30: (1-Tosyl-1H-indo-4-yl)methanol
Figure US10966961-20210406-C00039
Methyl 1-tosyl-1H-indole-4-carboxylate (6.248 g, 18.97 mmol) was taken up in DCM (200 mL) under nitrogen and cooled to −78° C. DIBAL-H (1M, 83 mL, 83 mmol) was added slowly over ˜30 min and the reaction left to stir at −78° C. for 90 min before being left to stir and warm to rt over the weekend. The reaction was cooled in an ice-bath and slowly quenched with MeOH (2 mL in 0.1 mL aliquots). Saturated Rochelle's salt solution (200 mL) was added and the mixture stirred and allowed to warm to rt for 3 h. The layers were separated and the organic layer eluted through a hydrophobic frit then concentrated in vacuo to give a yellow oil. The crude product was applied to a 100 g silica gel cartridge in the minimum of DCM and eluted with 5-50% ethyl acetate in cyclohexane. The appropriate fractions were concentrated in vacuo to give the desired product (5.172 g, 16.30 mmol, 86% yield) as a white solid.
LCMS (2 min High pH): Rt=1.07 min, [MH]+=301.2
Intermediate 31: Ethyl 1-tosyl-1H-pyrrolo[2,3-b]pyridine-4-carboxylate
Figure US10966961-20210406-C00040
Ethyl 1H-pyrrolo[2,3-b]pyridine-4-carboxylate (1.00 g, 5.26 mmol, commercially available from, for example, Alfa Aesar) was taken up in DMF (10 mL) and cooled in an ice bath. NaH (60% suspension in mineral oil, 0.252 g, 6.31 mmol) was added, and the reaction left to stir for 15 min. Tosyl chloride (1.203 g, 6.31 mmol) was added and the reaction was left to warm up to rt, and stirred for 1 h. The reaction was cooled in an ice bath, and quenched with water (5 mL). The reaction was concentrated in vacuo, brine (50 mL) was added to the residue and it was extracted with ethyl acetate (2×50 mL). This organic layer was dried with Na2SO4, filtered and concentrated in vacuo to yield a brown solid. The crude product was applied to a 50 g silica gel cartridge in the minimum of DCM and eluted with 5-25% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield a cream solid. This crude product was taken up in 1:2 MeOH:DCM (15 mL) and free flow silica added (10 g). The solvent was removed in vacuo and the silica loaded onto a 100 g ULTRA SNAP cartridge and eluted with 5-25% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were concentrated in vacuo to yield the desired product (1.084 g, 2.52 mmol, 48% yield) as a cream solid.
LCMS (2 min Formic): Rt=1.27 min, [MH]+=345.1
Intermediate 32: (1-Tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)methanol
Figure US10966961-20210406-C00041
Ethyl 1-tosyl-1H-pyrrolo[2,3-b]pyridine-4-carboxylate (936 mg, 2.72 mmol) was taken up in DCM (25 mL) under nitrogen and cooled to −78° C. DIBAL-H (1M in THF, 27.2 mL, 27.2 mmol) was added to the reaction dropwise and the reaction was left to stir at −78° C. overnight. The reaction was quenched with MeOH (1 mL added in small portions), and then allowed to warm to rt. Rochelle's salt (saturated in 50 mL of water) was added and the mixture was stirred for 15 min. The layers were separated and the organic layer was washed with brine (2×25 mL), dried with Na2SO4, filtered and concentrated in vacuo to yield an orange solid. The aqueous layers were extracted with 10% MeOH in DCM (50 mL). The two resulting organic layers were combined, dried with Na2SO4, filtered and concentrated in vacuo to yield an orange oil. The crude products were combined and applied to a 25 g silica gel cartridge in the minimum of DCM and eluted with 5-40% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were concentrated in vacuo to yield the desired product as a white solid (659 mg, 2.07 mmol, 76% yield).
LCMS (2 min Formic): Rt=0.92 min, [MH]+=303.1
Intermediate 33: (S)-Methyl 1-(1-(4-chlorophenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00042
Methyl 3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (0.5 g, 2.73 mmol) was taken up in THF (20 mL) and acetonitrile (5 mL). (R)-1-(4-Chlorophenyl)ethan-1-ol (0.4 mL, 2.96 mmol, commercially available from, for example, Sigma Aldrich) and Ph3P (1.078 g, 4.11 mmol) were added, and the reaction was put under nitrogen and cooled to 0° C. DIAD (0.8 mL, 4.11 mmol) was added dropwise and the reaction was allowed to warm to rt and left to stir for 90 min. EtOAc (25 mL) was added to the reaction mixture, followed by water (50 mL). The layers were separated and the organic layer washed with brine (50 mL), dried with Na2SO4, filtered and concentrated in vacuo to yield a yellow oil. The crude product was applied to a 100 g ULTRA SNAP cartridge in the minimum of DCM and eluted with 5-50% ethyl acetate in cyclohexane. The desired fractions were combined and concentrated in vacuo to yield methyl (S)-1-(1-(4-chlorophenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (415 mg, 1.225 mmol, 45% yield) as a colourless gum.
LCMS (2 min Formic): Rt=1.08 min, [MH]+=322.4.
The following intermediates were prepared in a similar manner to Intermediate 33.
LCMS:
Intermediate (System, tRET, MH+)
34
Figure US10966961-20210406-C00043
Formic, 1.04 min, 302.3
(S)-Methyl 3-(methylcarbamoyl)-1-(1-(m-
tolyl)ethyl)-1H-pyrazole-5-carboxylate
35
Figure US10966961-20210406-C00044
Formic, 1.07 min, 302.2
(S)-Methyl 3-(methylcarbamoyl)-1-(1-
phenylpropyl)-1H-pyrazole-5-carboxylate
36
Figure US10966961-20210406-C00045
Formic, 1.00 min, 306.1
(S)-Methyl 1-(1-(4-fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-pyrazole-5-
carboxylate
37
Figure US10966961-20210406-C00046
Formic, 0.99 min, 306.4
(S)-Methyl 1-(1-(3-fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-pyrazole-5-
carboxylate
38
Figure US10966961-20210406-C00047
Formic, 0.99 min, 306.4
(S)-Methyl 1-(1-(2-fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-pyrazole-5-
carboxylate
39
Figure US10966961-20210406-C00048
Formic, 1.18 min, 467.3
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-1H-
indol-4-yl)methyl)-1H-pyrazole-5-
carboxylate
40
Figure US10966961-20210406-C00049
Formic, 1.22 min, MH 479
Methyl 3-(methylcarbamoyl)-1-(1-(1-tosyl-
1H-indol-4-yl)ethyl)-1H-pyrazole-5-
carboxylate
41
Figure US10966961-20210406-C00050
Formic, 1.16 min, 467.5
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-1H-
indol-5-yl)methyl)-1H-pyrazole-5-
carboxylate
42
Figure US10966961-20210406-C00051
Formic, 1.23 min, [M − H]− at 479.5
Methyl 3-(methylcarbamoyl)-1-(1-(1-tosyl-
1H-indol-5-yl)ethyl)-1H-pyrazole-5-
carboxylate
Intermediate 43: (S)-Methyl 1-(1-(3-chlorophenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00052
Methyl 3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (0.5 g, 2.73 mmol), (R)-1-(3-chlorophenyl)ethan-1-01 (0.513 g, 3.28 mmol), cyanomethylenetributylphosphorane (CMBP) (1.432 mL, 5.46 mmol, commercially available from, for example, TCI) and toluene (10 mL) were added to the reaction vessel. The reaction vessel was sealed and heated in a Biotage Initiator microwave to 150° C. for 30 min. After cooling the reaction, it was concentrated in vacuo to yield a brown oil. The crude product was applied to a 100 g ULTRA SNAP cartridge in the minimum of DCM and eluted with 5-60% ethyl acetate in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield methyl (S)-1-(1-(3-chlorophenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (462 mg, 1.364 mmol, 50% yield) as an orange solid.
LCMS (2 min Formic): Rt=1.09 min, [MH]+=322.2.
The following intermediates were prepared in a similar manner to Intermediate 43.
LCMS:
Intermediate (System, tRET, MH+)
44
Figure US10966961-20210406-C00053
Formic, 0.83 min, 468.3
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-
1H-pyrrolo[2,3-c]pyridin-4-yl)methyl)-1H-
pyrazole-5-carboxylate
45
Figure US10966961-20210406-C00054
Formic, 1.11 min, 482.3
Methyl 3-(methylcarbamoyl)-1-(1-(1-tosyl-
1H-pyrrolo[2,3-b]pyridin-4-yl)ethyl)-1H-
pyrazole-5-carboxylate
46
Figure US10966961-20210406-C00055
Formic, 1.06 min, 468.2
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-
1H-pyrrolo[2,3-b]pyridin-4-yl)methyl)-1H-
pyrazole-5-carboxylate
47
Figure US10966961-20210406-C00056
Formic, 1.06 min, 322.3
(S)-Methyl 1-(1-(2-chlorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-pyrazole-5-
carboxylate
48
Figure US10966961-20210406-C00057
Formic, 0.99 min, 318.2
(S)-Methyl 1-(1-(3-methoxyphenyl)ethyl)-
3-(methylcarbamoyl)-1H-pyrazole-5-
carboxylate
Intermediate 49: 3-(Methylcarbamoyl)-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00058
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-5-carboxylate (265 mg, 0.426 mmol) was taken up in methanol (2.5 mL) and THF (2.5 mL). 1M LiOH in water (0.852 mL, 0.852 mmol) was added and the mixture stirred at 50° C. for 90 min. The reaction was concentrated in vacuo. The residue was partitioned between ethyl acetate and water (10 mL each). The aqueous layer was acidified with 2M HCl, and then extracted with ethyl acetate (2×10 mL), dried with Na2SO4, filtered and concentrated in vacuo to yield the desired product (173.5 mg, 0.326 mmol, 77% yield) as a white solid
LCMS (2 min Formic): Rt=1.13 min, [MH]+=453.3
The following intermediates were prepared in a similar manner to intermediate 49.
LC
In- MS:
ter- (Sys-
me- tem,
di- tRET,
ate Intermediate used MH+)
50
Figure US10966961-20210406-C00059
Figure US10966961-20210406-C00060
For- mic, 0.86 min, 274.2
(S)-3-(Methylcarbamoyl)-1-(1- Intermediate 34
(m-tolyl)ethyl)-1H-pyrazole-5-
carboxylic acid
51
Figure US10966961-20210406-C00061
Figure US10966961-20210406-C00062
For- mic, 0.96 min, 288.2
(S)-3-(Methylcarbamoyl)-1-(1- Intermediate 35
phenylpropyl)-1H-pyrazole-5-
carboxylic acid
52
Figure US10966961-20210406-C00063
Figure US10966961-20210406-C00064
For- mic, 0.88 min, 292.1
(S)-1-(1-(4- Intermediate 36
Fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
53
Figure US10966961-20210406-C00065
Figure US10966961-20210406-C00066
For- mic, 0.90 min, 292.4
(S)-1-(1-(3- Intermediate 37
Fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
54
Figure US10966961-20210406-C00067
Figure US10966961-20210406-C00068
For- mic, 0.89 min, 292.4
(S)-1-(1-(2- Intermediate 38
Fluorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
55
Figure US10966961-20210406-C00069
Figure US10966961-20210406-C00070
High pH, 0.90 min, 404.6
(R)-1-(2-((tert- Intermediate 90
Butyldimethylsilyl)oxy)-1-
phenylethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
56
Figure US10966961-20210406-C00071
Figure US10966961-20210406-C00072
For- mic, 1.16 min, MH 465
3-(Methylcarbamoyl)-1-(1-(1- Intermediate 40
tosyl-1H-indol-4-yl)ethyl)-1H-
pyrazole-5-carboxylic acid
57
Figure US10966961-20210406-C00073
Figure US10966961-20210406-C00074
For- mic, 1.06 min, MH 468
3-(Methylcarbamoyl)-1-(1-(1- Intermediate 45
tosyl-1H-pyrrolo[2,3-b]pyridin-
4-yl)ethyl)-1H-pyrazole-5-
carboxylic acid
58
Figure US10966961-20210406-C00075
Figure US10966961-20210406-C00076
For- mic, 1.11 min, 453.6
3-(Methylcarbamoyl)-1-((1- Intermediate 41
tosyl-1H-indol-5-yl)methyl)-1H-
pyrazole-5-carboxylic acid
59
Figure US10966961-20210406-C00077
Figure US10966961-20210406-C00078
For- mic, 1.17 min, [M − H]− at 465.5
3-(Methylcarbamoyl)-1-(1-(1- Intermediate 42
tosyl-1H-indol-5-yl)ethyl)-1H-
pyrazole-5-carboxylic acid
60
Figure US10966961-20210406-C00079
Figure US10966961-20210406-C00080
For- mic, 0.44 min, 300.1
1-((1H-Pyrrolo[2,3-b]pyridin-4- Intermediate 46
yl)methyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
61
Figure US10966961-20210406-C00081
Figure US10966961-20210406-C00082
For- mic, 0.99 min, 308.2
(S)-1-(1-(4- Intermediate 33
Chlorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
62
Figure US10966961-20210406-C00083
Figure US10966961-20210406-C00084
For- mic, 0.99 min, 308.1
(S)-1-(1-(3- Intermediate 43
Chlorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
63
Figure US10966961-20210406-C00085
Figure US10966961-20210406-C00086
For- mic, 0.95 min, 308.2
(S)-1-(1-(2- Intermediate 47
Chlorophenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
64
Figure US10966961-20210406-C00087
Figure US10966961-20210406-C00088
For- mic, 0.88 min, 304.1
(S)-1-(1-(3- Intermediate 48
Methoxyphenyl)ethyl)-3-
(methylcarbamoyl)-1H-
pyrazole-5-carboxylic acid
Intermediate 65: N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00089
3-(Methylcarbamoyl)-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-5-carboxylic acid (173 mg, 0.382 mmol) was taken up in DMF (5 mL). DIPEA (0.200 mL, 1.147 mmol), then HATU (218 mg, 0.573 mmol) were added and the reaction left to stir for 5 min, giving a brown solution. (1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-amine hydrochloride (For a preparation, see intermediate 94, 57.0 mg, 0.421 mmol) was then added, and the reaction was left to stir at rt overnight. The reaction mixture was partitioned between ethyl acetate and sodium bicarbonate (15 mL each). The organic layer was washed with 2M HCl (15 mL), brine (15 mL), and then dried with Na2SO4, filtered and concentrated in vacuo to give the crude product as a yellow oil. The crude product was purified by silica gel column chromatography, eluting with 10-60% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to give the desired product (99.7 mg, 0.178 mmol, 46% yield) as a cream solid.
LCMS (2 min Formic): Rt=1.09 min, [MH]+=534.3
Intermediate 66: 1-((1H-Indol-4-yl)methyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00090
3-(Methylcarbamoyl)-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-5-carboxylic acid (139 mg, 0.307 mmol) was taken up in THF (6 mL) and methanol (3 mL). Cesium carbonate (500 mg, 1.536 mmol) was added and the reaction was left to stir at 70° C. for 3.5 h. The reaction was concentrated in vacuo, water (10 mL) was added to the residue, which was then acidified with 2N HCl, and extracted with ethyl acetate (2×10 mL). This organic layer was dried with Na2SO4, filtered and concentrated in vacuo to yield the desired product (91.5 mg, 0.291 mmol, 95% yield) as a pink solid.
LCMS (2 min Formic): Rt=0.75 min, [MH]+=299.1
Intermediate 67: (1R,5S,6s)-tert-Butyl 6-(1-((1H-indol-4-yl)methyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxamido)-3-azabicyclo[3.1.0]hexane-3-carboxylate
Figure US10966961-20210406-C00091
1-((1H-indol-4-yl)methyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (78.6 mg, 0.263 mmol) was taken up in DMF (5 mL). HATU (150 mg, 0.395 mmol), then DIPEA (0.138 mL, 0.790 mmol) were added and the reaction was left to stir for 5 min, giving a brown solution. tert-Butyl (1R,5S,6s)-6-amino-3-azabicyclo[3.1.0]hexane-3-carboxylate, hydrochloride (68.0 mg, 0.290 mmol, commercially available from, for example, Astatech) was added and the reaction was left to stir at rt for 1 h. The reaction was concentrated in vacuo. The residue was partitioned between ethyl acetate and sodium bicarbonate (10 mL each). The layers were separated and the organic layer was washed with 2N HCl (10 mL), brine (10 mL), and then dried with Na2SO4, filtered and concentrated in vacuo to yield the crude product as a brown solid. The crude product was applied to a 10 g ULTRA SNAP cartridge in the minimum of DCM (with a couple of drops of MeOH) and eluted with 5-50% (3:1 EtOAc:EtOH) in cyclohexane. The appropriate fractions were combined and concentrated in vacuo to yield the desired product (93.8 mg, 0.186 mmol, 71% yield) as a cream solid.
LCMS (2 min Formic): Rt=1.00 min, [MH]+=479
Intermediate 68: 3-Hydroxybicyclo[3.1.0]hexane-6-carboxylic acid
Figure US10966961-20210406-C00092
Sodium hydroxide (6 mL, 12.00 mmol, 2M aqueous solution) was added to a solution of ethyl 3-hydroxybicyclo[3.1.0]hexane-6-carboxylate (1 g, 5.88 mmol, commercially available from, for example, Fluorochem) in ethanol (10 mL) and the mixture was stirred for 2 h at rt, then evaporated in vacuo to half its original volume and acidified with 2M HCl to ˜pH 2. The solution was saturated with sodium chloride (solid added and the mixture stirred for 1 h), then extracted first with 10% MeOH/DCM (5×10 mL) and then with EtOAc (5×10 mL). The combined organics were dried over sodium sulphate and evaporated in vacuo to give 3-hydroxybicyclo[3.1.0]hexane-6-carboxylic acid (0.65 g, 4.57 mmol, 78% yield) as a colourless gum. This was used crude in subsequent reactions.
Intermediate 69: 6-Aminobicyclo[3.1.0]hexan-3-ol
Figure US10966961-20210406-C00093
3-Hydroxybicyclo[3.1.0]hexane-6-carboxylic acid (650 mg, 4.57 mmol) was dissolved in toluene (10 mL) and tert-butanol (10 mL), then Et3N (1.275 mL, 9.15 mmol) and diphenyl phosphorazidate (1.478 mL, 6.86 mmol) were added and the mixture was heated at 80° C. overnight. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL), the solvent was dried and evaporated to give a pale yellow gum. This was dissolved in DCM (10 mL) and loaded onto a 50 g silica column, then eluted with 0-100% EtOAc/cyclohexane and ninhydrin active fractions were evaporated in vacuo to give three batches of impure Boc-protected amine intermediates (each likely with a different diastereomer as the major component), batch A (105 mg), batch B (122 mg) and batch C (85 mg). Each of these was dissolved in DCM (3 mL) and TFA (1 mL) was added, the solutions were stirred for 1 h, then evaporated in vacuo and the residue dissolved in methanol and loaded onto 5 g SCX cartridges. These were washed with methanol and then eluted with 2M methanolic ammonia. The eluants were evaporated in vacuo to give three batches of product as pale yellow glasses (each likely with a different diastereomer as the major component), batch A (15 mg), batch B (12 mg) and batch C (14 mg). The products were used crude and coupled in subsequent reactions.
Intermediate 70: tert-Butyl(cyclopent-3-en-1-yloxy)dimethylsilane
Figure US10966961-20210406-C00094
Cyclopent-3-en-1-ol (5 g, 59.4 mmol, commercially available from, for example, Astatech) was dissolved in DCM (100 mL) and TBDMS-Cl (8.96 g, 59.4 mmol) and imidazole (4.86 g, 71.3 mmol) were added, then the resulting suspension was stirred at rt over the weekend. The mixture was washed with water (2×100 mL), dried and evaporated in vacuo to give tert-butyl(cyclopent-3-en-1-yloxy)dimethylsilane (12.05 g, 60.7 mmol, 102% yield) as a pale yellow liquid.
1H NMR (400 MHz, CDCl3) δ ppm 5.68 (s, 2H) 4.50-4.62 (m, 1H) 2.59 (dd, J=14.9, 6.8 Hz, 2H) 2.23-2.37 (m, 2H) 0.91 (s, 9H) 0.09 (s, 6H).
Intermediate 71: (1R,5S,6r)-Ethyl 3-((tert-Butyldimethylsily)oxy)bicyclo[3.1.0]hexane-6-carboxylate, Mixture of Diastereomers
Figure US10966961-20210406-C00095
Ethyl diazoacetate (6.90 mL, 66.5 mmol) was dissolved in DCM (150 mL) and added dropwise over 5 h to a mixture of rhodium(II) acetate dimer (1 g, 2.263 mmol) and tert-butyl(cyclopent-3-en-1-yloxy)dimethylsilane (12 g, 60.5 mmol) in DCM (150 mL) at rt. The resulting green solution was stirred overnight, then evaporated in vacuo to give a green liquid. This was loaded onto a 340 g silica column and eluted with 0-40% EtOAc/cyclohexane and TLC plates of the fractions were visualised using permanganate dip. Active fractions were evaporated in vacuo to give ethyl (1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexane-6-carboxylate (5.5 g, 19.33 mmol, 32.0% yield) as a colourless liquid, as a mixture of isomers at the silyl ether position (˜3:1 ratio) and this was carried through crude to the next step.
LCMS (2 min High pH): Rt=0.96 min, [MI-1]±=not present.
Intermediate 72: (1R,5S,6r)-3-((tert-Butyldimethylsilyloxy)bicyclo[3.1.0]hexane-6-carboxylic acid, Mixture of Diastereomers
Figure US10966961-20210406-C00096
Sodium hydroxide (20 mL, 40.0 mmol, 2M aqueous solution) was added to a solution of ethyl (1R,5S,6r)-3-((tert-butyldimethylsilyloxy)bicyclo[3.1.0]hexane-6-carboxylate (5.0 g, 17.58 mmol) in ethanol (50 mL) at rt and the mixture was stirred for 3 h. TLC suggested that all the starting material had been consumed and the mixture was evaporated in vacuo to about 30 mL volume, then diluted with water (30 mL) and washed with ether (50 mL). The ether washings from the workup were dried and evaporated in vacuo to give recovered starting material:ethyl (1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexane-6-carboxylate (3.85 g). This was dissolved in ethanol (30 mL) and 2M aqueous NaOH solution (20 mL) was added, then the mixture was heated at 70° C. for 3 h, then evaporated in vacuo. The residue was dissolved in water (50 mL) and washed with ether (50 mL), then the aqueous layer was acidified with 2M HCl (20 mL) and extracted with EtOAc (2×50 mL). The combined organics were dried and evaporated in vacuo to give (1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexane-6-carboxylic acid (1.9 g, 7.41 mmol, 42.2% yield) as a pale yellow solid, the NMR is consistent with a mixture of isomers. The product was carried through to the next step without purification.
Intermediate 73: Benzyl ((1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-6-yl)carbamate, Mixture of Diastereomers
Figure US10966961-20210406-C00097
(1R,5S,6r)-3-((tert-Butyldimethylsilyl)oxy)bicyclo[3.1.0]hexane-6-carboxylic acid (1.8 g, 7.02 mmol) was dissolved in a mixture of toluene (20 mL) and Et3N (1.957 mL, 14.04 mmol), then DPPA (1.815 mL, 8.42 mmol) was added and the mixture was stirred for 30 min at rt. Benzyl alcohol (1.095 mL, 10.53 mmol) was added and the mixture heated at 100° C. for 4 h, then cooled to rt. Ethyl acetate (100 mL) was added and the solution was washed with water (2×100 mL), then dried over sodium sulphate, filtered and the filtrate evaporated in vacuo to give a pale yellow oil. This was dissolved in DCM (10 mL) and loaded onto a 50 g silica column, then eluted with 0-30% EtOAc/cyclohexane and product-containing fractions (detected by permanganate dip) were collected and evaporated in vacuo to give benzyl ((1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-6-yl)carbamate (1.90 g, 5.26 mmol, 75% yield) as a pale yellow oil, the NMR is consistent with desired product as a mixture of isomers in approximately 2:1 ratio. The compound was taken through to the next step without further purification.
LCMS (2 min Formic): Rt=1.56 min, [MH]+=362.6.
Intermediate 74: (1R,5S,6r)-3-((tert-Butyldimethylsilypoxy)bicyclo[3.1.0]hexan-6-amine, Mixture of Diastereomers
Figure US10966961-20210406-C00098
Benzyl ((1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-6-yl)carbamate (1.9 g, 5.26 mmol) was dissolved in ethanol (100 mL) and hydrogenated in the H-Cube at atmospheric pressure and 1 mL/min flow rate. The eluant was evaporated in vacuo to give (1R,5S,6r)-3-((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-6-amine (1.12 g, 4.92 mmol, 84% yield) as a pale yellow oil. Product is an unequal mixture of diastereomers at the silyl ether position with a ratio of approximately 65:35.
1H NMR (400 MHz, CDCl3) δ ppm 4.22 ppm, 1H [A](br.t, CH), 3.80 ppm 1H [B] (m, CH), 2.47 ppm, 1H [A] (t, CH), 2.05-1.93 ppm 2H [A]+3H [B] (m, 5×CH), 1.75-1.66 ppm NH2 [A]+—NH2 [B]+2×CH [B}, 1.62 ppm 2H, [A] (dd, 2×CH). Both diasteromers assigned. 1.20-1.15 ppm 2H [A]+2H [B] (M, 4×CH), 0.86 ppm, 9H [A] (s, 3×CH3)+9H [B] (s, 3×CH3), 0.00 ppm, 6H [A+B] (s, 2×CH3)
Intermediate 75: N5-((1r,4r)-4-Hydroxycyclohexyl)-N3-methyl-1-(1-(1-tosyl-1H-indol-4-yl)ethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00099
To a solution of 3-(methylcarbamoyl)-1-(1-(1-tosyl-1H-indol-4-yl)ethyl)-1H-pyrazole-5-carboxylic acid (255 mg, 0.547 mmol) in DMF (4 mL) was added HATU (312 mg, 0.820 mmol) followed by (1r,4r)-4-aminocyclohexan-1-ol (126 mg, 1.093 mmol, commercially available from, for example, Fluorochem) and DIPEA (0.477 mL, 2.73 mmol). The resulting reaction mixture was stirred at rt in air for 5 h. The reaction mixture was concentrated in vacuo to remove DMF and partitioned between ethyl acetate and saturated aq. LiCl solution. The organic layer was separated, washed with brine, dried (hydrophobic frit) and concentrated to give a crude orange oil. This was purified by chromatography on silica gel column, eluting with 0-50% (25% ethanol in ethyl acetate)/ethyl acetate to give the desired product (330 mg, 0.439 mmol, 80% yield) as a pale yellow oil.
LCMS (2 min Formic): Rt=1.07 min, [M+Na]+=586
The following intermediates were prepared in a similar manner to Intermediate 75:
Inter-
mediate
76
Figure US10966961-20210406-C00100
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-1-
((R)-2-((tert-
butyldimethylsilyl)oxy)-1-
phenylethyl)-N3-methyl-1H-
pyrazole-3,5-dicarboxamide
77
Figure US10966961-20210406-C00101
N3-Methyl-N5-((1S,2S)-2-
methylcyclopropyl)-1-(1-(1-
tosyl-1H-pyrrolo[2,3-b]pyridin-
4-yl)ethyl)-1H-pyrazole-3,5-
dicarboxamide
78
Figure US10966961-20210406-C00102
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-
N3-methyl-1-((1-tosyl-1H-indol-
5-yl)methyl)-1H-pyrazole-3,5-
dicarboxamide
79
Figure US10966961-20210406-C00103
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-
N3-methyl-1-(1-(1-tosyl-1H-
indol-5-yl)ethyl)-1H-pyrazole-
3,5-dicarboxamide
80
Figure US10966961-20210406-C00104
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-
N3-methyl-1-(1-(1-tosyl-1H-
indol-4-yl)ethyl)-1H-pyrazole-
3,5-dicarboxamide
81
Figure US10966961-20210406-C00105
N5-((1R*,5S*,6r*)-3-((tert-
Butyldimethylsilyl)oxy)bicyclo
[3.1.0]hexan-6-yl)-1-((S)-1-(3-
chlorophenyl)ethyl)-N3-methyl-
1H-pyrazole-3,5-dicarboxamide
82
Figure US10966961-20210406-C00106
N5-((1R,5S,6r)-3-((tert-
Butyldimethylsilyl)oxy)bicyclo
[3.1.0]hexan-6-yl)-N3-methyl-1-
((S)-1-phenylpropyl)-1H-
pyrazole-3,5-dicarboxamide
LCMS:
Inter- (System,
mediate Intermediate used tRET, MH+)
76
Figure US10966961-20210406-C00107
High pH, 1.29 min, 485.7
Intermediate 55
77
Figure US10966961-20210406-C00108
Formic, 1.11 min, 521.1
Intermediate 57
78
Figure US10966961-20210406-C00109
Formic, 1.06 min, 534.6
Intermediate 58
79
Figure US10966961-20210406-C00110
Formic, 1.12 min, [M − H]− at 546.5
Intermediate 59
80
Figure US10966961-20210406-C00111
Formic, 1.12 min, MH 546.4
Intermediate 56
81
Figure US10966961-20210406-C00112
Formic, 1.57 min, 517.6
Intermediate 62
82
Figure US10966961-20210406-C00113
High pH, 1.55 min, 497.7
Intermediate 51
Intermediate 83: 1-((1H-Pyrrolo[2,3-c]pyridin-4-yl)methyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00114
Methyl 3-(methylcarbamoyl)-1-((1-tosyl-1H-pyrrolo[2,3-c]pyridin-4-yl)methyl)-1H-pyrazole-5-carboxylate (106 mg, 0.227 mmol) was taken up in methanol (1.5 mL) and THF (1.5 mL). 1M LiOH in water (0.453 mL, 0.453 mmol) was added and the reaction left to stir at rt overnight. Additional 1M LiOH in water (0.453 mL) was added, and the reaction left to stir for 4 h. The residue was partitioned between water and ethyl acetate (10 mL of each). The aqueous layer was acidified with 2N HCl and extracted with ethyl acetate (2×15 mL). The aqueous layer was then extracted with 10% MeOH in DCM (2×15 mL), and the aqueous layer concentrated in vacuo to give the crude residue. The crude product was dissolved in 1:1 DMSO:MeCN (2 mL), filtered and then purified by MDAP (High pH). The appropriate fractions were concentrated in vacuo to yield the title compound (22 mg, 0.051 mmol, 23% yield) as a cream solid.
LCMS (2 min Formic): Rt=0.37 min, [MH]+=300.2
Intermediate 84: (S)—N5-(3,3-Diethoxypropyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00115
(S)-3-(Methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (121 mg, 0.443 mmol) was dissolved in DMF (2 mL). HATU (255 mg, 0.671 mmol) was added followed by 3,3-diethoxypropan-1-amine (0.12 mL, 0.742 mmol, commercially available from, for example, Acros) and then DIPEA (0.39 mL, 2.233 mmol). The reaction mixture was stirred under nitrogen for 3.25 h. The reaction mixture was partitioned between sat. aqueous LiCl (20 mL), Na2CO3 (˜1 mL), and ethyl acetate (20 mL); the layers were separated. The aqueous layer was extracted with further ethyl acetate (2×20 mL). The organic layers were combined, back extracted with water (2×10 mL) and filtered through a cartridge fitted with a hydrophobic frit. The filtrate was evaporated in vacuo to give a colourless gum. This was redissolved in dichloromethane and directly applied to the top of a 10 g SNAP silica cartridge and purified by SP4 flash column chromatography (0-60% ethyl acetate/cyclohexane). The relevant fractions were combined to give (S)—N5-(3,3-diethoxypropyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (103 mg, 0.230 mmol, 52% yield)
LCMS (2 min High pH): Rt=1.05 min, [MH]+=403.2.
Intermediate 85: 2-(1,3-Dihydroxypropan-2-yl)isoindoline-1,3-dione
Figure US10966961-20210406-C00116
To a solution of 2-aminopropane-1,3-diol (2.839 g, 31.2 mmol, commercially available from, for example, Sigma Aldrich) in DMF (60 mL) was added isobenzofuran-1,3-dione (4.62 g, 31.2 mmol, commercially available from, for example, Sigma Aldrich) dropwise at rt. The reaction was heated to 90° C. and stirred overnight. The reaction mixture was partitioned between EtOAc (200 mL) and water (250 mL) and the layers separated. The organic layer was analysed and shown to contain product. Therefore the aqueous layer was further extracted with EtOAc (2×100 mL) and then the combined organics were back extracted with water (2×50 mL) and sat. aq. LiCl solution (50 mL). The organics were then dried (Na2SO4) and concentrated in vacuo to afford 2-(1,3-dihydroxypropan-2-yl)isoindoline-1,3-dione (3.33 g, 15.05 mmol, 48% yield) as a cream solid.
LCMS (2 min High pH): Rt=0.52 min, [MH]+=222.2.
Intermediate 86: N5-(2-((2r,5S)-5-(1,3-Dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)ethyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00117
A mixture of (S)—N3-(3,3-diethoxypropyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (113 mg, 0.280 mmol), 2-(1,3-dihydroxypropan-2-yl)isoindoline-1,3-dione (62 mg, 0.280 mmol) and p-toluenesulfonic acid monohydrate (12 mg, 0.063 mmol) in toluene (3 mL) was stirred at 110° C. under nitrogen for 2 h. The reaction mixture was then cooled to rt and the volatiles evaporated in vacuo to give a yellow solid. This was partitioned between 10% methanol in DCM (10 mL), water (5 mL) and sat. aqueous Na2CO3 (10 mL) and the layers separated. The aqueous phase was extracted with 10% methanol in DCM (3×10 mL) and ethyl acetate (5 mL). The organic layers were combined and filtered through a cartridge fitted with a hydrophobic frit. The organic layers were concentrated in vacuo to give 138 mg of crude product. The crude product was taken up in DCM and purified by flash chromatography (10 g SNAP silica cartridge) eluting with 0-100% EtOAc/cyclohexane. The relevant fractions were combined and concentrated in vacuo to give N5-(2-((2r,5S)-5-(1,3-dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)ethyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (102 mg, 0.154 mmol, 55% yield).
LCMS (2 min High pH): Rt=1.10 min, [MH]+=532.2.
Intermediate 87: (S)—N5-(4,4-Diethoxybutyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00118
(S)-3-(Methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (125 mg, 0.457 mmol) was dissolved in DMF (2 mL). HATU (272 mg, 0.715 mmol) was added followed by 4,4-diethoxybutan-1-amine (0.13 mL, 0.752 mmol, commercially available from, for example, Sigma Aldrich) and then DIPEA (0.4 mL, 2.290 mmol). The reaction mixture was stirred under nitrogen for 6 h. Further HATU (209 mg, 0.549 mmol), 4,4-diethoxybutan-1-amine (0.1 mL, 0.579 mmol) and then DIPEA (0.1 mL, 0.573 mmol) were added before stirring for a further 3 h. The reaction mixture was partitioned between sat. aqueous LiCl (20 mL) and ethyl acetate (20 mL) and the layers separated. The aqueous layer was extracted with further ethyl acetate (2×20 mL). The organic layers were combined, back extracted with water (2×10 mL) and filtered through a cartridge fitted with a hydrophobic frit. The filtrate was evaporated in vacuo to give a colourless gum. This was redissolved in dichloromethane and directly applied to the top of a 10 g SNAP silica cartridge and purified by SP4 flash column chromatography (0-70% ethyl acetate/cyclohexane). The relevant fractions were combined and concentrated in vacuo to give (S)—N5-(4,4-diethoxybutyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (190 mg, 0.411 mmol, 90% yield).
LCMS (2 min High pH): Rt=1.07 min, [M−H]=415.3.
Intermediate 88: N5-(3-((2r,5S)-5-(1,3-Dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)propyl)-M-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00119
A mixture of (S)—N3-(4,4-diethoxybutyl)-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (190 mg, 0.456 mmol), 2-(1,3-dihydroxypropan-2-yl)isoindoline-1,3-dione (112 mg, 0.506 mmol) and p-toluenesulfonic acid monohydrate (19 mg, 0.100 mmol) in toluene (4 mL) was stirred at 110° C. under nitrogen for 2 h. The reaction mixture was then cooled to rt and the volatiles evaporated in vacuo to give a yellow solid. This was partitioned between 10% methanol in DCM (10 mL), water (5 mL) and sat. aqueous Na2CO3 (10 mL) and the layers separated. The aqueous phase was extracted with 10% methanol in DCM (3×10 mL) and ethyl acetate (5 mL) and the organic layers were combined and filtered through a cartridge fitted with a hydrophobic frit. The organic layers were concentrated in vacuo to give 207 mg of crude product. The crude product was taken up in DCM and purified by flash chromatography (25 g SNAP silica cartridge) eluting with 0-100% EtOAc/cyclohexane. The relevant fractions were combined and concentrated in vacuo to give N5-(3-((2r,5S)-5-(1,3-dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)propyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (81 mg, 0.119 mmol, 26% yield).
LCMS (2 min High pH): Rt=1.12 min, [MH]+=546.2.
Intermediate 89: (S)-2-((tert-Butyldimethylsilypoxy)-1-phenylethanol
Figure US10966961-20210406-C00120
(S)-1-Phenylethane-1,2-diol (251 mg, 1.817 mmol, commercially available from, for example, Sigma Aldrich) was dissolved in DCM (7 mL) before the addition of DMAP (22 mg, 0.180 mmol). The solution was cooled to 0° C. before the addition of triethylamine (0.380 mL, 2.72 mmol) and, dropwise, TBDMS-Cl (411 mg, 2.72 mmol) in DCM (3 mL). The reaction mixture was then stirred at rt for 2 h. To the reaction mixture was added NH4Cl (7 mL). The layers were separated and the aqueous was re-extracted with DCM (2×10 mL). The combined organic layers were dried and concentrated in vacuo to give 448 mg of crude product which was used directly in the next step.
LCMS (2 min High pH): Rt=1.41 min, [M−H]=251.1.
Intermediate 90: (R)-Methyl 1-(2-((tert-Butyldimethylsilypoxy)-1-phenylethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00121
Methyl 3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (216 mg, 1.179 mmol), (S)-2-((tert-butyldimethylsilyl)oxy)-1-phenylethan-1-ol (327 mg, 1.297 mmol) in toluene (2.5 mL), tri-n-butylphosphine (0.582 mL, 2.359 mmol) and 2-(tributylphosphoranylidene)acetonitrile (0.464 mL, 1.769 mmol) were combined. The reaction mixture was heated in a 5 mL microwave vial at 120° C. for 45 min. Further (S)-2-((tert-butyldimethylsilyl)oxy)-1-phenylethan-1-ol (119 mg, 0.471 mmol) was added to the reaction mixture before heating for 45 min at 120° C. in a microwave. The reaction mixture was concentrated in vacuo to give 1.574 g of crude product. The crude product was re-dissolved in DCM and purified by flash chromatography (100 g SNAP silica cartridge) eluting with 0-65% EtOAc:cyclohexane. The relevant fractions were combined and concentrated in vacuo to give methyl (R)-1-(2-((tert-butyldimethylsilyl)oxy)-1-phenylethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (276 mg, 0.562 mmol, 48% yield).
LCMS (2 min Formic): Rt=1.42 min, [MH]+=418.4.
Intermediate 91: (S)-3-(Methoxycarbonyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00122
LiOH (0.888 g, 21.16 mmol) in water (20 mL) was added to a solution of dimethyl (S)-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxylate (6.1 g, 21.16 mmol) in methanol (50 mL) and 2-MeTHF (50 mL) at 0° C. and the solution was stirred at 0° C. for 2 h, then acidified with 2M HCl (12 mL) and evaporated in vacuo to half the original volume. The mixture was further acidified to pH 2, then extracted with EtOAc (2×50 mL) and the combined organics dried and evaporated in vacuo to give a colourless gum. This was dissolved in DCM and loaded onto a 100 g silica column, then eluted with 0-50% (1% AcOH in EtOAc)/cyclohexane and the product-containing fractions were evaporated in vacuo to give (S)-3-(methoxycarbonyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (4.2 g, 15.31 mmol, 72% yield)
LCMS (2 min Formic): Rt=1.00 min, [MH]+=275.2.
Intermediate 92: (1R,5S,6r)-3-Oxabicyclo[3.1.0]hexane-6-carboxylic acid
Figure US10966961-20210406-C00123
LiOH (751 mg, 31.4 mmol) was added to a solution of (1R,5S,6r)-ethyl 3-oxabicyclo[3.1.0]hexane-6-carboxylate (1000 mg, 6.27 mmol, commercially available from, for example, Pharmablock) in water (10 mL), THF (10 mL) and MeOH (10 mL) at rt. The resulting suspension was stirred for 3 h. For work-up, the mixture was evaporated, the remaining crude solid was dissolved in a minimum amount of water, and quenched with HCl (5 mL, 25% m/m), and extracted 4 times with MeOH/DCM solvent, the combined organic phases were dried over a hydrophobic frit, evaporated in vacuo, to yield the desired compound (1R,5S,6r)-3-oxabicyclo[3.1.0]hexane-6-carboxylic acid (750 mg, 5.85 mmol, 93% yield) 1H NMR (400 MHz, DMSO-d6) δ ppm 12.13 (s, 1H) 3.80 (d, J=8.6 Hz, 2H) 3.62 (d, J=8.6 Hz, 2H) 2.00-2.15 (m, 2H) 1.32 (t, J=3.1 Hz, 1H)
Intermediate 93: Benzyl (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-ylcarbamate
Figure US10966961-20210406-C00124
(1R,5S,6r)-3-Oxabicyclo[3.1.0]hexane-6-carboxylic acid (340 mg, 2.65 mmol) was dissolved in toluene (12 mL), then Et3N (1.110 mL, 7.96 mmol), diphenyl phosphorazidate (0.686 mL, 3.18 mmol) and benzyl alcohol (0.552 mL, 5.31 mmol) were added and the mixture was heated at reflux for 2 h. The solution was diluted with EtOAc (10 mL) and washed with water (10 mL) and NaHCO3 solution (10 mL), the organic layer was dried and evaporated and the residue purified by chromatography on a 25 g silica column eluting with 0-50% EtOAc/cyclohexane and the product-containing fractions were evaporated in vacuo to give benzyl (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-ylcarbamate (460 mg, 1.972 mmol, 74.3% yield) as a white solid.
LCMS (2 min Formic): Rt=0.83 min, [MH]+=234.3.
1H NMR (400 MHz, CDCl3) δ ppm 7.29-7.41 (m, 5H) 5.11 (br. s., 2H) 4.86 (br. s., 1H) 3.98 (d, J=8.3 Hz, 2H) 3.72 (d, J=8.6 Hz, 2H) 2.45-2.52 (m, 1H) 1.80 (br. s, 2H) Intermediate 94: (1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-amine, hydrochloride
Figure US10966961-20210406-C00125
Benzyl (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-ylcarbamate (460 mg, 1.972 mmol) was dissolved in EtOH (20 mL) and the reaction was hydrogenated using an H-cube (settings: rt, 1 bar, 1 mL/min flow rate) and 10% Pd/C CatCart 30 as the catalyst. The reaction was cycled though the H-Cube for 1.5 h before acidifying the mixture with HCl (7M aqueous, 1.332 mL, 9.86 mmol) and evaporating in vacuo to yield an oily solid. The solid was dried in vacuo over 2 days to yield the desired product (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-amine, hydrochloride (262 mg, 1.836 mmol, 93% yield) as an off-white solid.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.48 (br. s., 3H) 3.80 (d, J=8.8 Hz, 2H) 3.59 (d, J=8.6 Hz, 2H) 2.24 (t, J=2.3 Hz, 1H) 2.07 (t, J=2.6 Hz, 2H).
Intermediate 95: 4-Methoxycyclopent-1-ene
Figure US10966961-20210406-C00126
Cyclopent-3-en-1-ol (10 g, 119 mmol, commercially available from, for example, Fluorochem) was dissolved in a mixture of DMF (100 mL) and THF (50 mL) and cooled in an ice bath under nitrogen, then NaH (60% suspension in mineral oil, 5.71 g, 143 mmol) was added in small portions and the mixture stirred for 30 min, before addition of MeI (9.66 mL, 155 mmol). The resulting suspension was stirred at 0° C. for 2 h, then added to water (500 mL) and extracted with ether (500 mL). The organic layer was washed with water (2×200 mL) and brine (200 mL), dried and evaporated in vacuo to give the desired product as a pale yellow liquid, which was carried through to subsequent reactions without further purification.
1H NMR (400 MHz, CDCl3) δ ppm 5.61-5.82 (m, 2H) 4.13 (dt, J=6.72, 3.48 Hz, 1H) 3.35 (s, 3H) 2.59 (dd, J=15.77, 6.72 Hz, 2H) 2.32-2.47 (m, 2H)
Intermediate 96: Ethyl 3-methoxybicyclo[3.1.0]hexane-6-carboxylate
Figure US10966961-20210406-C00127
Ethyl diazoacetate (82 mL, 119 mmol) was dissolved in DCM (100 mL) and added dropwise to a mixture of 4-methoxycyclopent-1-ene (For a preparation, see Intermediate 95, 11.68 g, 119 mmol) and rhodium(II) acetate dimer (2.63 g, 5.95 mmol) in DCM (100 mL) at rt over 4 h, then the mixture was stirred for a further 2 h, then washed with water (300 mL) and the organic layer dried and evaporated in vacuo to give a crude pale green liquid. This was dissolved in cyclohexane and loaded onto a 100 g SNAP ultra silica column and purified by flash chromatography, eluting with 0-20% EtOAc/cyclohexane. The appropriate fractions were evaporated in vacuo to give ethyl 3-methoxybicyclo[3.1.0]hexane-6-carboxylate (1.4 g, 7.60 mmol, 6% yield) as a colourless oil as a mixture of isomers also containing some residual ethyl diazoacetate. This was taken on for subsequent reactions without any further purification.
1H NMR (400 MHz, CDCl3) δ ppm 4.26 (q, J=7.09 Hz, 2H) 3.51 (m, J=7.50, 7.50 Hz, 1H) 3.25 (s, 3H) 2.26 (dd, J=13.08, 6.97 Hz, 2H) 1.89 (td, J=2.93, 1.22 Hz, 2H) 1.74-1.83 (m, 2H) 1.27-1.34 (m, 4H)
Intermediate 97: 3-Methoxybicyclo[3.1.0]hexane-6-carboxylic acid
Figure US10966961-20210406-C00128
Ethyl 3-methoxybicyclo[3.1.0]hexane-6-carboxylate (For a preparation, see Intermediate 96, 1.4 g, 7.60 mmol) was dissolved in ethanol (10 mL) and an aqueous solution of 2M NaOH (10 mL, 20.00 mmol) was added, then the mixture was stirred at rt overnight. The solvent was evaporated in vacuo and the residue was acidified with 2M HCl(aq.) (11 mL), then extracted with EtOAc (2×20 mL). The combined organics were dried and evaporated in vacuo to give a pale yellow oil which was used directly in the next step.
1H NMR (400 MHz, CDCl3) δ ppm 3.54 (m, J=7.50, 7.50 Hz, 1H) 3.28 (s, 3H) 2.30 (dd, J=13.20, 7.09 Hz, 2H) 1.99 (td, J=2.93, 1.22 Hz, 2H) 1.74-1.89 (m, 2H) 1.31 (t, J=2.93 Hz, 1H). Exchangeable proton not observed
Intermediate 98: tert-Butyl (3-methoxybicyclo[3.1.0]hexan-6-yl)carbamate
Figure US10966961-20210406-C00129
3-Methoxybicyclo[3.1.0]hexane-6-carboxylic acid (For a preparation, see Intermediate 97, 0.74 g, 4.74 mmol), diphenyl phosphoryl azide (1.956 g, 7.11 mmol) and Et3N (1.321 mL, 9.48 mmol) were dissolved in toluene (20 mL) and the mixture was stirred for 30 min, then tert-butanol (6 mL) was added and the mixture was heated at reflux for 3 h, then allowed to stand at rt overnight. The mixture was diluted with EtOAc (50 mL), then washed with water and sodium bicarbonate aqueous solution and evaporated in vacuo. The resulting gum was dissolved in DCM and loaded onto a 25 g SNAP ultra silica column, then purified by flash chromoatography eluting with 0-30% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give tert-butyl (3-methoxybicyclo[3.1.0]hexan-6-yl)carbamate (0.45 g, 1.980 mmol, 42% yield) as a colourless solid.
1H NMR (400 MHz, CDCl3) δ ppm 4.58 (br. s, 1H) 3.48 (quin, J=7.34 Hz, 1H) 3.26 (s, 3H) 2.24 (dd, J=12.84, 6.97 Hz, 2H) 2.16 (br. s., 1H) 1.71-1.80 (m, 2H) 1.39-1.49 (m, 11H)
Intermediate 99: 3-Methoxybicyclo[3.1.0]hexan-6-amine, hydrochloride
Figure US10966961-20210406-C00130
tert-Butyl (3-methoxybicyclo[3.1.0]hexan-6-yl)carbamate (For a preparation, see Intermediate 98, 380 mg, 1.672 mmol) was dissolved in HCl (4M in dioxane, 5 mL, 20.00 mmol) and the reaction mixture stirred at rt for 5 h. The reaction mixture was concentrated to give 3-methoxybicyclo[3.1.0]hexan-6-amine hydrochloride (270 mg, 1.402 mmol, 84% yield) as a light brown solid.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.20 (br. s., 3H) 3.54 (m, J=7.30 Hz, 1H) 3.16 (s, 3H) 2.25 (br. s., 1H) 2.05-2.14 (m, 2H) 1.60-1.70 (m, 4H) Intermediate 100: 7,7-Dimethyl-1,4-dioxaspiro[4.5]decan-8-one
Figure US10966961-20210406-C00131
To a solution of 1,4-dioxaspiro[4.5]decan-8-one (5.45 g, 34.9 mmol, commercially available from, for example, Sigma Aldrich) in dry THF (20 mL) was added sodium hydride (60% in mineral oil, 2.79 g, 69.8 mmol) at 0° C. under nitrogen. The resulting reaction mixture was stirred at rt for 1 h followed by the addition of methyl iodide (5.45 mL, 87 mmol). The reaction mixture was stirred at 10° C. to rt over 1.5 h. The reaction mixture was quenched with saturated ammonium chloride solution (50 mL) and the aqueous layer was extracted with ethyl acetate (2×80 mL). The combined organic layers were washed with brine and dried through a hydrophobic frit. The organic layer was concentrated in vacuo to give ˜6.8 g of crude yellow residue. This was purified by chromatography on SiO2 (Biotage SNAP Ultra 100 g cartridge, eluting with 5-65% diethyl ether/cyclohexane) to give 7,7-dimethyl-1,4-dioxaspiro[4.5]decan-8-one (3.07 g, 14.16 mmol, 41% yield) as a colourless oil.
LCMS (2 min Formic): Rt=0.78 min, [MH]+=185.0.
1H NMR (400 MHz, CDCl3) δ ppm 3.95-4.06 (m, 4H) 2.56-2.64 (m, 2H) 2.02 (d, J=1.26 Hz, 2H) 1.87-1.94 (m, 2H) 1.19 (s, 6H)
Intermediate 101: 7,7-Dimethyl-1,4-dioxaspiro[4.5]decan-8-ol
Figure US10966961-20210406-C00132
To a solution of 7,7-dimethyl-1,4-dioxaspiro[4.5]decan-8-one (For a preparation, see Intermediate 100, 3.04 g, 16.50 mmol) in methanol (10 mL) at 0° C. (ice-bath) was added sodium borohydride (0.624 g, 16.50 mmol) portionwise and the resulting reaction mixture was stirred at rt for 1 h. Water (10 mL) was added and the methanol removed in vacuo. The reaction mixture was partitioned between water and DCM. The organic layer was separated and the aqueous layer further extracted with DCM (2×20 mL). The combined organic layers were dried (hydrophobic frit) and concentrated under vacuum to give 7,7-dimethyl-1,4-dioxaspiro[4.5]decan-8-ol (2.87 g, 13.10 mmol, 79% yield) as a colourless oil.
LCMS (2 min Formic): Rt=0.62 min, [MH]+=187.1.
1H NMR (400 MHz, CDCl3) δ ppm 3.82-4.04 (m, 4H) 3.42 (ddd, J=8.31, 4.66, 3.40 Hz, 1H) 1.53-1.90 (m, 5H) 1.40-1.50 (m, 2H) 1.03 (br. s., 6H)
Intermediate 102: 4-Hydroxy-3,3-dimethylcyclohexanone
Figure US10966961-20210406-C00133
To a solution of 7,7-dimethyl-1,4-dioxaspiro[4.5]decan-8-ol (For a preparation, see Intermediate 101, 2.87 g, 15.41 mmol) in methanol (13 mL) was added HCl (2M aqueous solution, 30 mL, 60.0 mmol) and the reaction mixture stirred at rt overnight. The solvent was removed in vacuo and the residue neutralised (to ˜pH 7) with sat. aqueous NaHCO3 solution. The aqueous layer was extracted with ethyl acetate (2×40 mL) and the combined organic layers were dried (hydrophobic frit) and concentrated in vacuo to give 4-hydroxy-3,3-dimethylcyclohexan-1-one (2.18 g, 12.26 mmol, 80% yield) as a pale yellow oil.
LCMS (2 min Formic): Rt=0.54 min, [MH]+=143.0.
1H NMR (400 MHz, CDCl3) δ ppm 3.74 (dt, J=6.67, 3.46 Hz, 1H) 2.37-2.67 (m, 2H) 2.22-2.36 (m, 1H) 2.04-2.18 (m, 2H) 1.97 (dt, J=13.60, 6.80 Hz, 1H) 1.02 (br. s, 6H). Exchangeable proton not observed.
Intermediate 103: 4-(Benzylamino)-2,2-dimethylcyclohexanol, Mixture of Diastereomers
Figure US10966961-20210406-C00134
4-Hydroxy-3,3-dimethylcyclohexan-1-one (For a preparation, see Intermediate 102, 124 mg, 0.872 mmol) was dissolved in dichloromethane (2 mL). Phenylmethanamine (0.114 mL, 1.046 mmol) and acetic acid (0.050 mL, 0.872 mmol) were added and the reaction mixture was stirred under N2 at rt
After 3.5 h, sodium triacetoxyborohydride (222 mg, 1.046 mmol) was added and the reaction mixture stirred at rt overnight. The reaction mixture was quenched with sat. aqueous NaHCO3 solution (5 mL) and the organic layer was separated. The aqueous layer was further extracted with DCM (2×10 mL) and the combined organic layers were dried (Na2SO4) and conc. to give 270 mg of crude yellow oil. This was purified by chromatography on SiO2 (Biotage SNAP 25 g cartridge, eluting with 0-100% of (25% EtOH in ethyl acetate)/cyclohexane) to give a mixture of products. The desired fractions were concentrated to give 4-(benzylamino)-2,2-dimethylcyclohexanol (63 mg, 0.27 mmol, 26% yield) as a pale yellow oil.
LCMS (2 min Formic): Rt=0.38 min, [MH]+=234.2.
Intermediate 104: (+/−)-4-Amino-2,2-dimethylcyclohexanol, Mixture of Diastereomers
Figure US10966961-20210406-C00135
4-(Benzylamino)-2,2-dimethylcyclohexan-1-ol (For a preparation, see Intermediate 103, 63 mg, 0.270 mmol) was dissolved in ethyl acetate (6 mL), 10% palladium on carbon (40 mg) was added, and the mixture stirred under an atmosphere of hydrogen at rt for 21 h. The reaction mixture was filtered through celite to remove the catalyst and concentrated to give 4-amino-2,2-dimethylcyclohexan-1-ol (35 mg, 0.208 mmol, 77% yield) as a colourless oil as a racemic mixture of diastereomers. 1H NMR (400 MHz, CDCl3) δ ppm 3.32 (dd, J=11.58, 4.53 Hz, 1H) 2.78-2.92 (m, 2H) 1.25-1.93 (m, 18H) 1.06-1.18 (m, 1H) 0.96-1.05 (m, 6H) 0.88-0.95 (m, 6H).
Intermediate 105: Methyl 1-tosyl-1H-indole-7-carboxylate
Figure US10966961-20210406-C00136
Methyl 1H-indole-7-carboxylate (1000 mg, 5.71 mmol, commercially available from, for example, Apollo Scientific) was taken up in DMF (18 mL) and cooled to 0° C. under nitrogen. Sodium hydride (60% suspension in mineral oil, 287 mg, 7.18 mmol) was added in small portions and the reaction was left to stir for 15 min. Tosyl-Cl (1308 mg, 6.86 mmol) was added, and the reaction was allowed to warm to rt and left to stir for 16 h. The reaction mixture was cooled to 0 degrees before the addition of further sodium hydride (60% suspension in mineral oil, 114 mg, 2.85 mmol). The reaction mixture was stirred for 15 min before being warmed to rt. The reaction mixture was stirred for a further 15 min before the addition of a further portion of tosyl-Cl (552 mg, 2.90 mmol). The resulting solution was stirred for 2 h. The reaction mixture was then heated to 60° C. for 1.5 h. The reaction mixture was quenched with water (˜6 mL) before being partitioned between sat. aqueous LiCl (100 mL) and ethyl acetate (30 mL). The layers were separated and the aqueous was extracted twice more with ethyl acetate (2×30 mL). The combined organic layers were dried (hydrophobic frit) and concentrated in vacuo to give the crude product as a yellow oil. The crude product was redissolved in dichloromethane and directly applied to the top of a 100 g SNAP silica cartridge and purified by SP4 flash column chromatography (0-25% ethyl acetate/cyclohexane). The relevant fractions were combined and concentrated in vacuo to give methyl 1-tosyl-1H-indole-7-carboxylate (1025 mg, 2.80 mmol, 49% yield).
LCMS (2 min High pH): Rt=1.18 min, [MH]+=330.1.
Intermediate 106: (1-Tosyl-1H-indol-7-yl)methanol
Figure US10966961-20210406-C00137
A solution of methyl 1-tosyl-1H-indole-7-carboxylate (For a preparation, see Intermediate 105, 1025 mg, 3.11 mmol) in dichloromethane (31 mL) under nitrogen, was cooled to −78° C. and DIBAL-H (1M solution in THF, 7.78 mL, 7.78 mmol) was added dropwise over 15 min and the reaction was stirred at −78° C. for 1.5 h. The reaction was left to stir for 18 h before the addition of further DIBAL-H (25% weight solution in toluene, 4.19 mL, 6.22 mmol). The reaction mixture was stirred for 20 h. The reaction was quenched with methanol when still at −78° C. and after allowed to warm to rt. The reaction was diluted with Rochelle's salt solution (10 mL) and stirred for 64 h. The layers were separated, and the aqueous phase was extracted with DCM (3×20 mL). The combined organic layers were dried through a hydrophobic frit, then concentrated in vacuo to yield the crude product (1-tosyl-1H-indol-7-yl)methanol (950 mg, 2.84 mmol, 91% yield) as an orange oil which was used as is for subsequent reactions.
LCMS (2 min High pH): Rt=1.18 min, [M-OH]+=284.1.
Intermediate 107: Methyl 1-tosyl-1H-indazole-4-carboxylate
Figure US10966961-20210406-C00138
Methyl 1H-indazole-4-carboxylate (530 mg, 3.01 mmol, commercially available from, for example, Sigma Aldrich) was taken up in DMF (6.4 mL) and cooled to 0° C. under nitrogen. Sodium hydride (60% suspension in mineral oil, 241 mg, 6.02 mmol) was added in small portions and the reaction was left to stir for 10 min. Tosyl-Cl (775 mg, 4.07 mmol) was added before stirring for 30 min at 0° C. The reaction was allowed to warm to rt and left to stir for 1 h. The reaction mixture was poured onto 250 mL of water before filtering. The precipitate was dried in a vacuum oven overnight to give methyl 1-tosyl-1H-indazole-4-carboxylate (719 mg, 1.959 mmol, 65% yield).
LCMS (2 min High pH): Rt=1.22 min, [MH]+=331.1.
Intermediate 108: (1-Tosyl-1H-indazol-4-yl)methanol
Figure US10966961-20210406-C00139
A solution of methyl 1-tosyl-1/indazole-4-carboxylate (For a preparation, see Intermediate 107, 804 mg, 2.434 mmol) in dichloromethane (24 mL) under nitrogen, was cooled to −78° C. and DIBAL-H (1.637 mL, 2.434 mmol, 25% wt in toluene) was added drop-wise over 15 min and the reaction was stirred at −78° C. for 62 h. Further DIBAL-H (3.27 mL, 4.87 mmol, 25% wt in toluene) was added to the reaction at −78° C. before stirring for 1 h. The reaction was quenched with methanol when still at −78° C. and after allowed to warm to rt. The reaction was diluted with Rochelle's salt solution (10 mL) and stirred for 16 h. The layers were separated, and the aqueous phase was extracted with DCM (3×20 mL). The combined organic layers were dried through a hydrophobic frit, then evaporated in vacuo to give (1-tosyl-1H-indazol-4-yl)methanol (686 mg, 2.042 mmol, 84% yield).
LCMS (2 min High pH): Rt=1.00 min, [MH]+=303.1.
Intermediate 109: (+/−)-1-(3-Fluoro-2-methylphenyl)ethanol
Figure US10966961-20210406-C00140
1-(3-Fluoro-2-methylphenyl)ethan-1-one (1000 mg, 6.57 mmol, commercially available from, for example, Alfa Aesar) was taken up in THF (10 mL) and ethanol (10 mL), cooled to 0° C., and put under nitrogen. Sodium borohydride (456 mg, 12.05 mmol) was added, and the reaction left to stir at rt for 1.5 h. The reaction was slowly quenched with 1M HCl until pH 3, before the addition of water. The layers were separated and the aqueous layer extracted twice more with DCM (2×20 mL). The organic layers were back extracted with sodium bicarbonate aq. solution, passed through a hydrophobic frit and concentrated in vacuo to yield a colourless oil as the crude product, 1-(3-fluoro-2-methylphenyl)ethan-1-ol (875 mg, 5.11 mmol, 78% yield).
LCMS (2 min Formic): Rt=0.85 min, No [MH]+
Intermediate 110: Ethyl 3-(benzyloxy)bicyclo[3.1.0]hexane-6-carboxylate, Mixture of Diastereomers
Figure US10966961-20210406-C00141
Ethyl diazoacetate (15 mL, 21.69 mmol) was dissolved in DCM (100 mL) and added dropwise to a mixture of ((cyclopent-3-en-1-yloxy)methyl)benzene (3.1 g, 17.79 mmol, commercially available from, for example, Fluorochem) and rhodium(II) acetate dimer (0.393 g, 0.890 mmol) in DCM (100 mL) at rt over 4 h, then the mixture was stirred for a further 2 h, then washed with water (300 mL) and the organic layer dried and evaporated in vacuo to give a pale green liquid. This was purified using silica gel column chromatography eluting with a gradient of 0-20% EtOAc/cyclohexane to give (+/−)-ethyl 3-(benzyloxy)bicyclo[3.1.0]hexane-6-carboxylate (0.91 g, 3.50 mmol, 20% yield) as a colourless liquid.
LCMS (2 min High pH): Rt=1.26 min, [MH]+=261.3
Intermediate 111: 3-(Benzyloxy)bicyclo[3.1.0]hexane-6-carboxylic acid, Mixture of Diastereomers
Figure US10966961-20210406-C00142
(+/−)-Ethyl 3-(benzyloxy)bicyclo[3.1.0]hexane-6-carboxylate (For a preparation, see Intermediate 110, 0.9 g, 3.46 mmol) was dissolved in EtOH (20 mL) and NaOH (5 mL, 10.00 mmol, 2M aqueous) was added, then the mixture was stirred at rt overnight. The solvent was evaporated in vacuo to about half its original volume, then acidified with 2M HCl (aq) to pH 4 and extracted with EtOAc (50 mL). The organic layer was dried and evaporated in vacuo to give (+/−)-3-(benzyloxy)bicyclo[3.1.0]hexane-6-carboxylic acid (0.74 g, 3.19 mmol, 92% yield) as a colourless solid and a mixture of diastereomers.
LCMS (2 min High pH): Rt=0.56 min, [MH]+=233.3
Intermediate 112: tert-Butyl (3-(benzyloxy)bicyclo[3.1.0]hexan-6-yl)carbamate, mixture of diastereomers
Figure US10966961-20210406-C00143
3-(Benzyloxy)bicyclo[3.1.0]hexane-6-carboxylic acid (For a preparation, see Intermediate 111, 0.74 g, 3.19 mmol), diphenyl phosphoryl azide (1.140 g, 4.14 mmol) and Et3N (0.888 mL, 6.37 mmol) were dissolved in toluene (20 mL) and the mixture was stirred for 30 min, then tert-butanol (6 mL) was added and the mixture was heated at reflux for 3 h, then allowed to stand at rt overnight. The mixture was diluted with EtOAc, then washed with water and sat NaHCO3 (aq.) and evaporated in vacuo. The resulting gum was purified using silica gel column chromatography eluting with a gradient of 0-30% EtOAc:cyclohexane to give tert-butyl ((1R,3r,5S,6s)-3-(benzyloxy)bicyclo[3.1.0]hexan-6-yl)carbamate (0.35 g, 1.154 mmol, 36.2% yield) as a colourless solid and a mixture of diastereomers.
LCMS (2 min High pH): Rt=1.26 min, [MH]+=304.3.
Intermediate 113: 3-(Benzyloxy)bicyclo[3.1.0]hexan-6-amine, HCl, Mixture of Diastereomers
Figure US10966961-20210406-C00144
tert-Butyl (3-(benzyloxy)bicyclo[3.1.0]hexan-6-yl)carbamate (For a preparation, see Intermediate 112, 350 mg, 1.154 mmol) was dissolved in HCl (4M in dioxane, 5 mL, 20.00 mmol) and reaction mixture stirred at rt for 4 h. The reaction mixture was concentrated to give 3-(benzyloxy)bicyclo[3.1.0]hexan-6-amine, hydrochloride (253 mg, 0.897 mmol, 78% yield) as a pale yellow solid and a mixture of diastereomers.
LCMS (2 min High pH): Rt=0.90 min, [MH]+=204.2.
Intermediate 114: (+/−)-7-Methyl-1,4-dioxaspiro[4.5]decan-8-one
Figure US10966961-20210406-C00145
To a solution of 1,4-dioxaspiro[4.5]decan-8-one (1.04 g, 6.66 mmol, available from commercial suppliers such as Apollo Scientific) in THF (67 mL) under N2 at −78° C. was added 1M LiHMDS in THF (7.32 mL, 7.32 mmol). The reaction was stirred for 1 h and MeI (0.547 mL, 8.66 mmol) was added dropwise. The reaction was stirred for 3 h at −78° C., and then left to warm up to rt overnight. It was then quenched with a sat. NH4Cl (aq) solution and extracted with EtOAc. The combined organics were filtered through a hydrophobic frit and concentrated in vacuo to a brown oil. This oil was purified by silica gel column chromatography eluting with a gradient of 0 to 32% EtOAc:cyclohexane to give (+/−)-7-methyl-1,4-dioxaspiro[4.5]decan-8-one (443 mg, 2.60 mmol, 39% yield) as a white solid.
1H NMR (400 MHz, MeOH-d4) δ ppm 3.95-4.16 (m, 4H) 2.58-2.88 (m, 2H) 2.24-2.41 (m, 1H) 2.04-2.17 (m, 2H) 1.90-2.01 (m, 1H) 1.71 (t, J=13.1 Hz, 1H) 0.94-1.07 (m, 3H)
Intermediate 115: (+/−)-(trans)-7-Methyl-1,4-dioxaspiro[4.5]decan-8-ol
Figure US10966961-20210406-C00146
To a solution of 7-methyl-1,4-dioxaspiro[4.5]decan-8-one (For a preparation, see Intermediate 114, 438 mg, 2.57 mmol) in THF (10 mL) at −78° C. was added a 2M LiAlH4 solution in THF (1.67 mL, 3.35 mmol). The mixture was stirred 1.5 h at −78° C. It was then allowed to warm to 0° C., the reaction mixture was carefully quenched with a Rochelle salt solution. EtOAc was added and the layers were separated, the aqueous layer was extracted with EtOAc and the combined organics were filtered through a hydrophobic frit and concentrated in vacuo to give (+/−)-(trans)-7-methyl-1,4-dioxaspiro[4.5]decan-8-ol (430 mg, 2.397 mmol, 93% yield) as a colourless oil.
1H NMR (600 MHz, DMSO-d6) δ=4.43 (d, J=5.5 Hz, 1H), 3.88-3.75 (m, 4H), 2.96 (ddt, J=4.8, 5.7, 10.1 Hz, 1H), 1.75-1.68 (m, 1H), 1.64-1.60 (m, 1H), 1.63-1.59 (m, 1H), 1.51-1.44 (m, 1H), 1.48-1.41 (m, 1H), 1.43-1.35 (m, 1H), 1.19 (t, J=12.9 Hz, 1H), 0.89 (d, J=6.6 Hz, 3H)
Intermediate 116: (+/−)-(trans)-4-Hydroxy-3-methylcyclohexanone
Figure US10966961-20210406-C00147
To a solution of (+/−)-(trans)-7-methyl-1,4-dioxaspiro[4.5]decan-8-ol (For a preparation, see Intermediate 115, 425 mg, 2.468 mmol) in acetone (7 mL) was added H2SO4 (18 mL, 9.00 mmol, 0.5M) and the solution was stirred for 20 h at rt. The reaction mixture was diluted with water and extracted with EtOAc. The combined organics were filtered through a hydrophobic frit and concentrated in vacuo to give (+/−)-(trans)-4-hydroxy-3-methylcyclohexan-1-one (290 mg, 2.265 mmol, 92% yield) as a yellow oil.
1H NMR (400 MHz, MeOH-d4) δ ppm 3.93 (d, J=2.3 Hz, 1H) 3.10 (td, J=10.2, 4.4 Hz, 1H) 1.90-2.13 (m, 2H) 1.73-1.86 (m, 1H) 1.25-1.59 (m, 2H) 1.09 (t, J=13.1 Hz, 1H) 0.97-1.03 (m, 3H).
Exchangeable Proton not Observed
Intermediate 117: (S)—N-((1S,3R,4R)-4-Hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide and (S)—N-((1S,3S,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide (1:1 Diasteromeric Mixture)
Figure US10966961-20210406-C00148
(+/−)-(trans)-4-hydroxy-3-methylcyclohexan-1-one (For a preparation, see Intermediate 116, 1.99 g, 15.53 mmol) was added to a solution of (R)-2-methylpropane-2-sulfinamide (2.304 g, 18.63 mmol) and tetraethoxytitanium (5.43 mL, 25.9 mmol) in THF (30 mL) and the reaction was stirred at 60° C. for 1.5 h and then at 70° C. for 1.5 h. The mixture was cooled to rt and then to −78° C. A 1M solution of L-selectride in THF (38.8 mL, 38.8 mmol) was added dropwise and the reaction mixture was allowed to warm slowly to rt with stirring overnight. The reaction mixture was then cooled to 0° C. and MeOH was added dropwise. The crude reaction mixture was poured onto brine. The resulting suspension was filtered through a plug of Celite, and the filter cake was washed with EtOAc. The filtrate was then concentrated in vacuo, diluted with water and extracted EtOAc. The combined organics were filtered through a hydrophobic frit and concentrated in vacuo to a yellow oil. This oil was purified using silica gel column chromatography eluting with a gradient of 10 to 42% of (25% EtOH in AcOEt):cyclohexane to give (S)—N-((1S,3R,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide and (S)—N-((1S,3S,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide (1:1 diasteromeric mixture) (303.5 mg, 1.301 mmol, 8% yield) as a yellow gum which was used directly in the next reaction.
Intermediate 118: (1R,2R,45)-4-Amino-2-methylcyclohexanol and (1S,2S,4S)-4-amino-2-methylcyclohexanol (1:1 Diastereomeric Mixture)
Figure US10966961-20210406-C00149
4M HCl in dioxane (1 mL, 4.00 mmol) was added to a solution of (S)—N-(1S,3R,4R)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide compound and (S)—N-((1S,3S,4S)-4-hydroxy-3-methylcyclohexyl)-2-methylpropane-2-sulfinamide (1:1 diasteromeric mixture, For a preparation, see Intermediate 117) (300 mg, 1.286 mmol) in DCM (5 mL) and the reaction was stirred overnight at rt. More 4M HCl in dioxane (1 mL, 4.00 mmol) was added and the reaction was stirred 6 h at rt. Further 4M HCl in dioxane (1 mL, 4.00 mmol) was added and the reaction was stirred 1 h at rt. The reaction was concentrated in vacuo, dissolved in MeOH and eluted through a 2 g SCX column (pre-conditioned with MeOH) washing with MeOH and 2M NH3 in MeOH solution. The ammonia fractions were concentrated in vacuo to give (1R,2R,4S)-4-amino-2-methylcyclohexanol and (1S,2S,4S)-4-amino-2-methylcyclohexanol (1:1 diastereomeric mixture) (74.5 mg, 0.577 mmol, 45% yield) as a brown gum.
1H NMR (400 MHz, DMSO-d6) δ ppm 4.17-4.51 (m, 2H) 2.91-3.03 (m, 2H) 2.86 (td, J=10.2, 4.3 Hz, 2H) 1.61-1.82 (m, 4H) 1.35-1.58 (m, 4H) 1.09-1.28 (m, 4H) 0.95-1.07 (m, 2H) 0.90 (dd, J=9.4, 6.7 Hz, 6H) 0.61-0.84 (m, 2H)
Intermediate 119: (R)-1-(3-Fluoro-4-methylphenyl)ethanol
Figure US10966961-20210406-C00150
(+)-DIP-Cl (1.265 g, 3.94 mmol) was taken up in THF (20 mL) and was cooled to −25° C. under N2. 1-(3-Fluoro-4-methylphenyl)ethan-1-one (0.5 g, 3.29 mmol, available from commercial suppliers such as Alfa Aesar) in THF (10 mL) was added and the reaction was left to stir overnight as it warmed from −25° C. to rt. The reaction was cooled to −35° C. and additional (+)-DIP-Cl (1.265 g, 3.94 mmol) in THF (5 mL) was added. The reaction was kept between −35 and −25° C. as it stirred for 6 h and then was allowed to warm to rt overnight. The reaction was quenched with acetaldehyde (0.5 mL, 8.85 mmol) and concentrated in vacuo. The residue was taken up in Et2O (20 mL) and diethanolamine (1.036 g, 9.86 mmol) was added, this was left to stir for 2 h. The white precipitate was filtered off through Celite, and the filtrate was concentrated in vacuo to give a colourless oil. This oil was purified using silica gel column chromatography eluting with a gradient of 0-25% EtOAc:cyclohexane to give (R)-1-(3-fluoro-4-methylphenyl)ethan-1-ol (836 mg, 2.71 mmol, 83% yield) as a colourless liquid.
LCMS (2 min Formic): Rt=0.89 min, no [MH]+
Intermediate 120: (+/−)-7-Chlorobicyclo[3.2.0]hept-2-en-6-one
Figure US10966961-20210406-C00151
Zinc (8.42 g, 129 mmol) was added in small portions over 1 h to a solution of (+/−)-7,7-dichlorobicyclo[3.2.0]hept-2-en-6-one (24 g, 136 mmol, available from commercial suppliers such as Alfa Aesar) in AcOH (100 mL) at rt. The mixture was stirred for a further 1 h, then diluted with water and extracted with diethyl ether. The organic layer was washed with water and sat. sodium bicarbonate solution and brine, then dried and evaporated in vacuo to give (+/−)-7-chlorobicyclo[3.2.0]hept-2-en-6-one (20 g, 140 mmol, purity=70%) as a pale yellow liquid.
1H NMR (400 MHz, CDCl3) δ ppm 5.89-5.97 (m, 1H) 5.70-5.79 (m, 1H) 5.03-5.14 (m, 1H) 3.78-3.97 (m, 2H) 2.66-2.80 (m, 1H) 2.41-2.55 (m, 1H)
Intermediate 121: (+/−)-Bicyclo[3.1.0]hex-2-ene-6-carboxylic acid, Mixture of Diastereomers
Figure US10966961-20210406-C00152
To a stirred mixture of KOH (2.16 g, 38.5 mmol) in 1,4-dioxane (24 mL) and water (10 mL) was added (+/−)-7-chlorobicyclo[3.2.0]hept-2-en-6-one (For a preparation, see Intermediate 120, 1.307 g, 5.50 mmol) dropwise. The flask containing the chloro-SM was then washed into the reaction vessel with further 1,4-dioxane (300 μL). The reaction was stirred for 30 min. The reaction was acidified with 2M HCl(aq.) to pH 2. The organics were extracted with DCM (3×40 mL) and the combined organics allowed to stand overnight. After 16 h, the combined organics were dried (Na2SO4), filtered and concentrated in vacuo to afford a brown oil. This was dried further in vacuo to afford the desired product as a brown waxy solid-(+/−)-bicyclo[3.1.0]hex-2-ene-6-carboxylic acid (581 mg, 4.68 mmol, 85% yield) which was a mixture of ˜40%-endo/˜60%-exo (desired).
LCMS (2 min Formic): (exo) Rt=0.65 min, no m/z; (endo) Rt=0.54 min, no m/z
Intermediate 122: (+/−)-Benzyl (cis)-bicyclo[3.1.0]hex-2-en-6-ylcarbamate
Figure US10966961-20210406-C00153
DPPA (26.0 mL, 121 mmol) was added to a solution of (+/−)-bicyclo[3.1.0]hex-2-ene-6-carboxylic acid, mixture of diastereomers (For a preparation, see Intermediate 121, 10 g, 81 mmol) and Et3N (22.46 mL, 161 mmol) in toluene (10 mL) at rt and the mixture was stirred for 30 min, then benzyl alcohol (16.75 mL, 161 mmol) was added and the mixture heated at reflux for 3 h. The solution was diluted with EtOAc and washed with water and sat. sodium bicarbonate solution (aq), then dried and evaporated in vacuo to give a brown oil. This oil was purified using silica gel column chromatography eluting with a gradient of 0-50% EtOAc: cyclohexane to give (+/−)-benzyl ((cis)-bicyclo[3.1.0]hex-2-en-6-yl)carbamate (3.7 g, 16.14 mmol, 20.03% yield) as a colourless solid.
LCMS (2 min Formic): Rt=1.08 min, [MH]+=230.3
Intermediate 123: (+/−)-Benzyl ((cis)-2-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate
Figure US10966961-20210406-C00154
Borane-methyl sulfide complex (2.299 mL, 24.21 mmol) was added to a solution of (+/−)-benzyl ((cis)-bicyclo[3.1.0]hex-2-en-6-yl)carbamate (For a preparation, see Intermediate 122, 3.7 g, 16.14 mmol) in THF (10 mL) at 0° C. and the mixture was stirred for 2 h, then allowed to warm to rt over 30 min. Water (2 mL) was added followed by aqueous solution of 2M NaOH (16.14 mL, 32.3 mmol). The mixture was cooled in an ice bath then H2O2 (30%, 3.30 mL, 32.3 mmol) was added and the mixture was stirred for a further 1 h. The mixture was diluted with EtOAc and water and the mixture stirred vigorously for 30 min, then the organic layer separated and the aqueous extracted with EtOAc. The combined organics were washed with 5% sodium thiosulphate solution (aq.) then dried and evaporated in vacuo to give a gum. This gum was triturated with ether and the resulting colourless solid collected by filtration. The filtrate was evaporated in vacuo to give a colourless gum which was purified by silica gel column chromatography eluting with a gradient of 0-50% EtOAc:cyclohexane. Later running fractions gave a colourless gum (0.53 g), which partially solidified on standing. This material was dissolved in EtOAc and allowed to stand overnight, giving a colourless solid, which was collected by filtration. The filtrate was evaporated in vacuo to give a colourless gum. This gum was purified using silica gel column chromatography eluting with a gradient of 0-50% EtOAc:cyclohexane to give (+/−) benzyl ((cis)-2-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate (180 mg, 0.728 mmol, 5% yield) as a colourless gum.
1H NMR (600 MHz, DMSO-d6) δ ppm 7.19-7.44 (m, 6H) 4.96-5.04 (m, 2H) 4.53 (d, J=1.0 Hz, 1H) 4.07 (t, J=4.6 Hz, 1H) 2.09 (br. s., 1H) 1.83 (m, J=1.0, 1.0 Hz, 1H) 1.62 (dd, J=12.1, 8.4 Hz, 1H) 1.32-1.40 (m, 3H) 1.09-1.23 (m, 1H)
Intermediate 124: (+/−)-(cis)-6-Aminobicyclo[3.1.0]hexan-2-ol
Figure US10966961-20210406-C00155
(+/−)-Benzyl ((cis)-2-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate (For a preparation, see Intermediate 123, 180 mg, 0.728 mmol) was dissolved in EtOH (10 mL) and 10% Pd/C (35 mg, 0.329 mmol) was added. The reaction mixture was hydrogenated under atmospheric pressure for 6.5 h. The catalyst was filtered off and the reaction mixture concentrated to give (+/−)-(cis)-6-aminobicyclo[3.1.0]hexan-2-01 (82 mg, 0.728 mmol, purity=80%) as a pale yellow oil which was used crude directly in the next step.
Intermediate 125: (+/−)-(trans)-3-Methoxycyclopentanamine hydrochloride
Figure US10966961-20210406-C00156
(+/−)-tert-butyl ((trans)-3-methoxycyclopentyl)carbamate (For a preparation, see Intermediate 126, 280 mg, 1.040 mmol) was taken up in a solution of 4M hydrochloric acid in dioxane (2.5 mL, 10.00 mmol) and was stirred for 2 h. The reaction mixture was concentrated in vacuo to give (+/−)-(trans)-3-methoxycyclopentan-1-amine hydrochloride (167 mg, 0.991 mmol, 95% yield).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.05 (br s, 3H) 3.85-3.89 (m, 1H) 3.50-3.57 (m, 1H) 3.17 (s, 3H) 1.89-2.04 (m, 3H) 1.70-1.77 (m, 1H) 1.55-1.65 (m, 2H).
Intermediate 126: (+/−)-tert-Butyl ((trans)-3-methoxycyclopentyl)carbamate
Figure US10966961-20210406-C00157
Crushed 3 Å molecular sieves were placed under vacuum in a 250 mL round-bottom flask and heated with a heat gun. The flask was allowed to cool, and (+/−)-tert-butyl ((trans)-3-hydroxycyclopentyl)carbamate (370 mg, 1.838 mmol, commercially available from, for example, Fluorochem) in anhydrous dichloromethane (13 mL) was added. N1,N1,N8,N8-Tetramethylnaphthalene-1,8-diamine (1.357 g, 6.33 mmol) was added, and the reaction was cooled to 0° C. and placed under an atmosphere of nitrogen. Trimethyloxonium tetrafluoroborate (680 mg, 4.60 mmol) was added, and the reaction allowed to warm to rt and stirred rapidly for 20 h. The reaction was diluted with dichloromethane (50 mL) and washed with water (50 mL). The aqueous layer was extracted with dichloromethane (2×25 mL), and the combined organics were washed with aqueous 0.5 M HCl solution (15 mL), saturated sodium bicarbonate solution (20 mL), and brine (30 mL). The organic phase was passed through a hydrophobic frit and concentrated in vacuo to yield a yellow oil. The crude product was redissolved in dichloromethane and directly applied to the top of a 25 g SNAP silica cartridge and purified by SP4 flash column chromatography eluting with a gradient of 0-100% ethyl acetate in cyclohexane. The relevant fractions were combined and concentrated in vacuo to give (+/−)-tert-butyl ((trans)-3-methoxycyclopentyl)carbamate (280 mg, 1.040 mmol, 57% yield).
1H NMR (400 MHz, DMSO-d6) δ ppm 6.80 (br d, 1H) 3.84 (q, 1H) 3.75-3.80 (m, 1H) 3.14 (s, 3H) 1.80-1.90 (m, 3H) 1.46-1.56 (m, 2H) 1.38 (s, 9H) 1.29-1.36 (m, 1H).
Intermediate 127: 1-(3-(2-Hydroxyethoxy)phenyl)ethanone
Figure US10966961-20210406-C00158
3-Hydroxyacetophenone (5.0 g, 36.7 mmol, commercially available from, for example, Sigma Aldrich), ethylene carbonate (4.85 g, 55.1 mmol) and potassium carbonate (5.08 g, 36.7 mmol) were mixed in DMF (50 mL) and heated at 120° C. overnight under nitrogen, then the mixture was diluted with water (200 mL) and extracted with EtOAc (2×100 mL). The combined organics were washed with water (200 mL) and brine (200 mL), dried and evaporated in vacuo to give a pale yellow oil. This was dissolved in DCM and loaded onto a 100 g SNAP Ultra column and eluted with 0-100% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give 1-(3-(2-hydroxyethoxy)phenyl)ethan-1-one (5.5 g, 30.5 mmol, 83% yield), which was used directly in the next step.
LCMS (2 min High pH): Rt=0.67 min, [M−H]=179.1.
Intermediate 128: 1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethanone
Figure US10966961-20210406-C00159
TBDMS-Cl (4.97 g, 33.0 mmol) was added to a mixture of 1-(3-(2-hydroxyethoxy)phenyl)ethan-1-one (For a preparation, see Intermediate 127, 5.4 g, 30.0 mmol) and imidazole (2.45 g, 36.0 mmol) in DCM (50 mL) and the mixture was stirred for 2 h, then allowed to stand over the weekend. The resulting suspension was washed with water (2×100 mL) and the organic layer dried and evaporated in vacuo to give a colourless oil. This was loaded onto a 100 g SNAP ultra silica column and purified by flash chromatography eluting with 0-30% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethan-1-one (6.6 g, 22.41 mmol, 75% yield) as a colourless liquid which was used directly in the next step.
LCMS (2 min Formic): Rt=1.46 min, [MH]+=295.3.
Intermediate 129: (+/−)-1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)-ethanol
Figure US10966961-20210406-C00160
Sodium borohydride (1.27 g, 33.6 mmol) was added to a solution of 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethan-1-one (For a preparation, see Intermediate 128, 6.6 g, 22.41 mmol) in ethanol (50 mL) at 0° C. and the mixture was stirred for 2 h, then quenched by very cautious addition of ammonium chloride solution (50 mL, initially slowly and dropwise-vigorous effervescence!). The resulting mixture was diluted with brine (50 mL) and extracted with EtOAc (2×100 mL), the combined organics dried and evaporated in vacuo to give 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethan-1-ol (5.61 g, 18.92 mmol, 84% yield) as a colourless oil which was used directly in the next step.
LCMS (2 min High pH): Rt=1.41 min, [MH]+=296.3.
Intermediate 130: (+/−)-Methyl 1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate
Figure US10966961-20210406-C00161
DIAD (1.38 mL, 7.10 mmol) in DCM (10 mL) was added dropwise over 5 min to a solution of methyl 3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (For a preparation, see Intermediate 1, 1 g, 5.46 mmol), 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)-ethan-1-01 (For a preparation, see Intermediate 129, 1.942 g, 6.55 mmol) and triphenylphosphine (1.862 g, 7.10 mmol) in DCM (20 mL) at 0° C. and the mixture was stirred overnight, allowing it to warm to rt. The solution was washed with water (20 mL), dried and evaporated in vacuo to give a pale yellow gum. This was dissolved in DCM and loaded onto a 100 g SNAP ultra silica column, then eluted with 0-60% EtOAc/cyclohexane and the product-containing fractions were evaporated in vacuo to give (+/−)-methyl 1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (2.61 g, 5.65 mmol, 104% yield) as a pale yellow gum, which was a 4:1 mixture of diastereomers (with alternate pyrazole alkylation product as minor component).
LCMS (2 min Formic): Rt=1.43 min, [MH]+=462.3.
Intermediate 131: (+/−)-1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid
Figure US10966961-20210406-C00162
(+/−)-Methyl 1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylate (For a preparation, see Intermediate 130, 2.6 g, 5.63 mmol) was dissolved in methanol (30 mL) and NaOH (10 mL, 20.00 mmol, 2M in water) was added, then the mixture was stirred at rt for 2 h. The solvent was then evaporated in vacuo. The resulting solid was dissolved in water (30 mL) and washed with ether (2×30 mL) then the aqueous layer was acidified with 2M HCl(aq.) (11 mL) and the resulting mixture was extracted with EtOAc (2×30 mL). The combined organics were washed with water, dried and evaporated in vacuo to give (+/−)-1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (2.22 g, 4.96 mmol, 88% yield) as a colourless gum, as an approximately 4:1 ratio of regioisomers (with alternate pyrazole alkylation product as minor component from previous step), which appeared to be inseparable at this stage.
LCMS (2 min High pH): Rt=0.99 min, [MH]+=448.4.
Intermediate 132: 1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-N3-methyl-N5-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide, Mixture of Diastereomers
Figure US10966961-20210406-C00163
(+/−)-1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (For a preparation, see Intermediate 131, 2 g, 4.47 mmol), (1S,2S)-2-methylcyclopropan-1-amine, hydrochloride (0.625 g, 5.81 mmol), HATU (2.209 g, 5.81 mmol) and Et3N (1.868 mL, 13.40 mmol) were dissolved in DCM (20 mL) and the mixture was stirred for 2 h, then allowed to stand over the weekend at rt. The resulting mixture was stirred with water (50 mL) for 1 h, then the organic layer was separated, dried and evaporated in vacuo to give a pale yellow gum. This was dissolved in DCM and loaded onto a 100 g SNAP ultra silica column and purified by flash chromatography eluting with 0-100% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give 1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-N3-methyl-N5-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide (1.10 g, 2.197 mmol, 49% yield, mixture of diastereomers) as a colourless solid.
LCMS (2 min Formic): Rt=1.44 min, [MH]+=501.4.
EXAMPLES Example 1: N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00164
To a solution of (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (98 mg, 0.359 mmol) in DMF (0.8 mL) was added HATU (205 mg, 0.538 mmol) followed by (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-amine, hydrochloride (107 mg, 0.789 mmol) and DIPEA (0.313 mL, 1.793 mmol). The resulting reaction mixture was stirred at rt in air for 18 h. The reaction mixture was purified by MDAP (Formic). The fractions containing desired product were partitioned between sat. NaHCO3 solution and DCM. The organic layer was separated and aqueous layer further extracted with DCM (2×20 mL). Combined organic layers were dried (Na2SO4) and concentrated in vacuo to give N-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (25 mg, 0.063 mmol, 18% yield) as a white solid.
1H NMR (400 MHz, MeOH-d4) δ ppm 7.20-7.32 (m, 5H) 7.09 (s, 1H) 6.59 (d, J=7.1 Hz, 1H) 3.99 (dd, J=8.3, 2.7 Hz, 2H) 3.72 (dt, J=8.2, 3.5 Hz, 2H) 2.93 (s, 3H) 2.53 (t, J=2.4 Hz, 1H) 1.92 (d, J=7.1 Hz, 3H) 1.85-1.90 (m, 1H) 1.77-1.82 (m, 1H). 2 exchangeable protons not observed.
LCMS (2 min Formic): Rt=0.85 min, [MH]+=355.3.
The crude product remaining after the first MDAP injection was also purified by MDAP (Formic) with fractions containing desired product partitioned between sat. NaHCO3 solution and DCM. The organic layer was separated and aqueous layer further extracted with DCM (2×20 mL). Combined organic layers were dried (Na2SO4) and concentrated in vacuo to give a second batch of N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (30 mg, 0.076 mmol, 21% yield) as a white solid.
LCMS (2 min Formic): Rt=0.85 min, [MH]+=355.3.
Example 2: N5-((1r,4.5)-4-Hydroxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00165
To a solution of (5)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (451 mg, 1.650 mmol) in DMF (5 mL) was added HATU (941 mg, 2.475 mmol) followed by (1r,4r)-4-aminocyclohexanol (trans) (380 mg, 3.30 mmol) and DIPEA (1.441 mL, 8.25 mmol). The resulting reaction mixture was stirred at rt in air for 40 min. The reaction mixture was concentrated in vacuo to remove DMF and partitioned between ethyl acetate and saturated aq. LiCl solution. The organic layer was separated, washed with brine, dried (hydrophobic frit) and concentrated to give ˜1.5 g of crude product as an orange oil (containing DMF). This was purified by chromatography on SiO2 (Biotage SNAP 25 g cartridge, eluting with 20-100% ethyl acetate/cyclohexane, followed by 100% ethyl acetate to 12% ethanol/ethyl acetate) to give 447 mg of a colourless oil. This was further purified by MDAP (Formic). Fractions containing the desired product were partitioned between sat. NaHCO3 solution and DCM. The organic layer was separated and aqueous layer further extracted with DCM (2×20 mL). Combined organic layers were dried (Na2SO4) and concentrated in vacuo to give N5-((1r,45)-4-hydroxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (163 mg, 0.396 mmol, 24% yield) as a white solid.
1H NMR (600 MHz, DMSO-d6) δ ppm 1.16-1.25 (m, 2H) 1.25-1.36 (m, 2H) 1.67-1.80 (m, 2H) 1.78-1.86 (m, 2H) 1.83 (d, J=6.0 Hz, 3H) 2.77 (d, J=4.8 Hz, 3H) 3.31-3.39 (m, 1H) 3.57-3.66 (m, 1H) 4.52 (d, J=4.5 Hz, 1H) 6.67 (q, J=7.0 Hz, 1H) 7.22-7.25 (m, 1H) 7.23-7.24 (m, 1H) 7.24-7.27 (m, 2H) 7.28-7.31 (m, 2H) 8.11 (q, J=4.5 Hz, 1H) 8.28 (d, J=7.8 Hz, 1H)
LCMS (2 min Formic): Rt=0.82 min, [MH]+=371.3.
Example 3: N5-((1R,3R,5S,6r)-3-Hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00166
Crude tert-butyl (3-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate (37 mg, 0.173 mmol) was dissolved in DCM (10 mL) and TFA (1 mL) was added, then the mixture was stirred for 2 h at rt. The solvent was evaporated in vacuo to give a pale yellow gum. The crude residue was dissolved in DCM (5 mL), then HATU (86 mg, 0.226 mmol), Et3N (0.048 mL, 0.347 mmol) and (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (47.4 mg, 0.173 mmol) were added and the mixture was stirred for 2 h at rt. The solvent was evaporated in vacuo and the residue purified by MDAP (high pH) to give N5-((1R,3R,5S,6s)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((5)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (3 mg, 8.14 μmol, 5% yield).
1H NMR (600 MHz, DMSO-d6) δ ppm 1.36-1.40 (m, 2H) 1.58 (ddt, J=12.4, 8.3, 4.4, 4.4 Hz, 2H) 1.82 (d, J=7.0 Hz, 3H) 2.01 (dt, J=12.6, 7.1 Hz, 2H) 2.42 (dt, J=3.9, 2.0 Hz, 1H) 2.75 (d, J=4.6 Hz, 3H) 3.80 (br t, J=7.0 Hz, 1H) 4.59 (br d, J=3.3 Hz, 1H) 6.68 (q, J=7.0 Hz, 1H) 7.19 (s, 1H) 7.21-7.26 (m, 1H) 7.23-7.26 (m, 2H) 7.27-7.33 (m, 2H) 8.13 (q, J=4.6 Hz, 1H) 8.42 (d, J=4.0 Hz, 1H)
LCMS (2 min High pH): Rt=0.79 min, [MH]+=369.4.
The following examples were prepared in a similar manner to Example 3:
LCMS:
Exam- (System,
ple Intermediate used tRET, MH+)
4
Figure US10966961-20210406-C00167
Figure US10966961-20210406-C00168
Formic, 0.88 min, 385.4
N5-((1r,4S)-4- Intermediate 50
Hydroxycyclohexyl)-N3-
methyl-1-((S)-1-(m-
tolyl)ethyl)-1H-pyrazole-3,5-
dicarboxamide
5
Figure US10966961-20210406-C00169
Figure US10966961-20210406-C00170
Formic, 0.94 min, 369.3
N5-((1R,5S,6r)-3- Intermediate 51
Oxabicyclo[3.1.0]hexan-6-yl)-
N3-methyl-1-((S)-1-
phenylpropyl)-1H-pyrazole-
3,5-dicarboxamide
6
Figure US10966961-20210406-C00171
Figure US10966961-20210406-C00172
Formic, 0.89 min, 385.3
N5-((1r,4S)-4- Intermediate 51
Hydroxycyclohexyl)-N3-
methyl-1-((S)-1-
phenylpropyl)-1H-pyrazole-
3,5-dicarboxamide
7
Figure US10966961-20210406-C00173
Figure US10966961-20210406-C00174
Formic, 0.87 min, 373.2
N3-((1R,5S,6r)-3- Intermediate 52
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(4-
fluorophenyl)ethyl)-N5-
methyl-1H-pyrazole-3,5-
dicarboxamide
8
Figure US10966961-20210406-C00175
Figure US10966961-20210406-C00176
Formic, 0.87 min, 373.5
N5-((1R,5S,6r)-3- Intermediate 53
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(3-
fluorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
9
Figure US10966961-20210406-C00177
Figure US10966961-20210406-C00178
Formic, 0.86 min, 373.6
N5-((1R,5S,6r)-3- Intermediate 54
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(2-
fluorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
10
Figure US10966961-20210406-C00179
Figure US10966961-20210406-C00180
High pH, 0.91 min, 313.3
(S)-N5-Cyclopropyl-N3-methyl- Intermediate 5
1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
11
Figure US10966961-20210406-C00181
Figure US10966961-20210406-C00182
Formic, 0.98 min, 327.3
N3-Methyl-N5-((1S,2S)-2- Intermediate 5
methylcyclopropyl)-1-((S)-1-
phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
12
Figure US10966961-20210406-C00183
Figure US10966961-20210406-C00184
Formic, 1.11 min, 454
(1R,5S,6s)-tert-Butyl 6-(3- Intermediate 5
(methylcarbamoyl)-1-((S)-1-
phenylethyl)-1H-pyrazole-5-
carboxamido)-3-
azabicyclo[3.1.0]hexane-3-
carboxylate
13
Figure US10966961-20210406-C00185
Figure US10966961-20210406-C00186
High pH, 0.70 min, 353.5
1-((1H-Pyrrolo[2,3-c]pyridin- Intermediate 83
4-yl)methyl)-N3-methyl-N5-
((1S,2S)-2-
methylcyclopropyl)-1H-
pyrazole-3,5-dicarboxamide
14
Figure US10966961-20210406-C00187
Figure US10966961-20210406-C00188
Formic, 0.60 min, 353.2
1-((1H-Pyrrolo[2,3-b]pyridin- Intermediate 60
4-yl)methyl)-N3-methyl-N5-
((1S,2S)-2-
methylcyclopropyl)-1H-
pyrazole-3,5-dicarboxamide
15
Figure US10966961-20210406-C00189
Figure US10966961-20210406-C00190
Formic, 0.97 min, 389.3
N5-((1R,5S,6r)-3- Intermediate 61
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(4-
chlorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
16
Figure US10966961-20210406-C00191
Figure US10966961-20210406-C00192
Formic, 0.92 min, 405.4
1-((S)-1-(4- Intermediate 61
Chlorophenyl)ethyl)-N5-
((1r,4S)-4-
hydroxycyclohexyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
17
Figure US10966961-20210406-C00193
Figure US10966961-20210406-C00194
Formic, 0.89 min, 405.1
1-((S)-1-(3- Intermediate 62
Chlorophenyl)ethyl)-N5-
((1r,4S)-4-
hydroxycyclohexyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
18
Figure US10966961-20210406-C00195
Figure US10966961-20210406-C00196
Formic, 0.94 min, 389.1
N5-((1R,5S,6r)-3- Intermediate 62
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(3-
chlorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
19
Figure US10966961-20210406-C00197
Figure US10966961-20210406-C00198
Formic, 0.87 min, 371.1
N5-(3-Hydroxycyclohexyl)-N3- Intermediate 5
methyl-1-((S)-1-phenylethyl)-
1H-pyrazole-3,5-
dicarboxamide, mixture of
diastereomers
20
Figure US10966961-20210406-C00199
Figure US10966961-20210406-C00200
Formic, 0.83 min, 339.0
(S)-N3-Methyl-1-(1- Intermediate 5
phenylethyl)-N5-(1H-pyrazol-
4-yl)-1H-pyrazole-3,5-
dicarboxamide
21
Figure US10966961-20210406-C00201
Figure US10966961-20210406-C00202
Formic, 0.86 min, 357.1
N5-((1S,3R)-3- Intermediate 5
Hydroxycyclopentyl)-N3-
methyl-1-((S)-1-phenylethyl)-
1H-pyrazole-3,5-
dicarboxamide
22
Figure US10966961-20210406-C00203
Figure US10966961-20210406-C00204
High pH, 0.93 min, 403.6
1-((S)-1-(4- Intermediate 61
Chlorophenyl)ethyl)-N5-
((1R,3S,5S,6r)-3-
hydroxybicyclo[3.1.0]hexan-6-
yl)-N3-methyl-1H-pyrazole-
3,5-dicarboxamide
23
Figure US10966961-20210406-C00205
Figure US10966961-20210406-C00206
Formic, 0.90 min, 373.5
(S)-1-(1-(3- Intermediate 62
Chlorophenyl)ethyl)-N3-
methyl-N5-(1H-pyrazol-4-yl)-
1H-pyrazole-3,5-
dicarboxamide
24
Figure US10966961-20210406-C00207
Figure US10966961-20210406-C00208
Formic, 0.91 min, 373.5
(S)-1-(1-(4- Intermediate 61
Chlorophenyl)ethyl)-N3-
methyl-N5-(1H-pyrazol-4-yl)-
1H-pyrazole-3,5-
dicarboxamide
25
Figure US10966961-20210406-C00209
Figure US10966961-20210406-C00210
High pH, 0.90 min, 403.6
1-((S)-1-(4- Intermediate 61
Chlorophenyl)ethyl)-N5-
((1R,3R,5S,6r)-3-
hydroxybicyclo[3.1.0]hexan-6-
yl)-N3-methyl-1H-pyrazole-
3,5-dicarboxamide
26
Figure US10966961-20210406-C00211
Figure US10966961-20210406-C00212
Formic, 0.61 min, 350.1
(S)-N3-Methyl-1-(1- Intermediate 5
phenylethyl)-N5-(pyridin-4-yl)-
1H-pyrazole-3,5-
dicarboxamide
27
Figure US10966961-20210406-C00213
Figure US10966961-20210406-C00214
Formic, 1.04 min, 329.1
N5-((R)-sec-Butyl)-N3-methyl- Intermediate 5
1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
28
Figure US10966961-20210406-C00215
Figure US10966961-20210406-C00216
Formic, 1.07 min, 341.1
(S)-N5-Cyclopentyl-N3-methyl- Intermediate 5
1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
29
Figure US10966961-20210406-C00217
Figure US10966961-20210406-C00218
Formic, 1.04 min, 329.2
N5-(sec-Butyl)-N3-methyl-1- Intermediate 5
((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide,
mixture of diastereomers
30
Figure US10966961-20210406-C00219
Figure US10966961-20210406-C00220
Formic, 1.06 min, 329.1
(S)-N5-Isobutyl-N3-methyl-1- Intermediate 5
(1-phenylethyl)-1H-pyrazole-
3,5-dicarboxamide
31
Figure US10966961-20210406-C00221
Figure US10966961-20210406-C00222
Formic, 0.91 min, 357.1
N3-Methyl-1-((S)-1- Intermediate 5
phenylethyl)-N5-(tetrahydro-
2H-pyran-3-yl)-1H-pyrazole-
3,5-dicarboxamide, mixture of
diastereomers
32
Figure US10966961-20210406-C00223
Figure US10966961-20210406-C00224
Formic, 1.02 min, 327.1
(S)-N5-Cyclobutyl-N3-methyl- Intermediate 5
1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
33
Figure US10966961-20210406-C00225
Figure US10966961-20210406-C00226
Formic, 0.85 min, 340.1
(S)-N3-Methyl-N5-(oxazol-2- Intermediate 5
yl)-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
34
Figure US10966961-20210406-C00227
Figure US10966961-20210406-C00228
Formic, 0.90 min, 345.1
(S)-N5-(3-Methoxypropyl)-N3- Intermediate 5
methyl-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
35
Figure US10966961-20210406-C00229
Figure US10966961-20210406-C00230
Formic, 0.89 min, 353.1
(S)-N3-Methyl-N5-(1-methyl- Intermediate 5
1H-pyrazol-4-yl)-1-(1-
phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
36
Figure US10966961-20210406-C00231
Figure US10966961-20210406-C00232
Formic, 0.80 min, 331.1
N5-(2-Hydroxypropyl)-N3- Intermediate 5
methyl-1-((S)-1-phenylethyl)-
1H-pyrazole-3,5-
dicarboxamide, mixture of
diastereomers
37
Figure US10966961-20210406-C00233
Figure US10966961-20210406-C00234
Formic, 0.90 min, 371.1
(S)-N3-Methyl-1-(1- Intermediate 5
phenylethyl)-N5-((tetrahydro-
2H-pyran-4-yl)methyl)-1H-
pyrazole-3,5-dicarboxamide
38
Figure US10966961-20210406-C00235
Figure US10966961-20210406-C00236
Formic, 0.55 min, 400.2
N3-Methyl-N5-(2-(4- Intermediate 5
methylmorpholin-2-yl)ethyl)-
1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide,
mixture of diastereomers
39
Figure US10966961-20210406-C00237
Figure US10966961-20210406-C00238
Formic, 0.77 min, 331.1
(S)-N5-(3-Hydroxypropyl)-N3- Intermediate 5
methyl-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
40
Figure US10966961-20210406-C00239
Figure US10966961-20210406-C00240
Formic, 0.90 min, 389.5
N5-((1R,5S,6r)-3- Intermediate 63
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(2-
chlorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
41
Figure US10966961-20210406-C00241
Figure US10966961-20210406-C00242
Formic, 0.87 min, 449.4
1-((S)-1-(2- Intermediate 63
Chlorophenyl)ethyl)-N5-
((1r,4S)-4-
hydroxycyclohexyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
42
Figure US10966961-20210406-C00243
Figure US10966961-20210406-C00244
Formic, 0.82 min, 385.2
N5-((1r,4S)-4-Hydroxy-4- Intermediate 5
methylcyclohexyl)-N3-methyl-
1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
43
Figure US10966961-20210406-C00245
Figure US10966961-20210406-C00246
Formic, 0.86 min, 385.3
N5-((1R,5S,6r)-3- Intermediate 64
Oxabicyclo[3.1.0]hexan-6-yl)-
1-((S)-1-(3-
methoxyphenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-
dicarboxamide
Examples 44-49 were prepared via an array with the following method:—
To a 20 mL vial was weighed (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (0.027 g, 0.1 mmol)×14=383 mg and HATU (0.042 g, 0.110 mmol)×14=586 mg. To this vial was added DMF (0.5 mL)×14=7 mL and DIPEA (0.050 mL, 0.286 mmol)×14=700 μL. The mixture was stirred for 5 min and then aliquoted into vials (550 μL) each containing the appropriate pre-weighed amine (0.120 mmol). (NOTE: additional DIPEA (0.050 mL, 0.286 mmol) was added to amines which were HCl salts). The mixture was allowed to react for 66 h. T3P (100 μL, 50 wt % in EtOAc) and DIPEA (0.050 mL, 0.286 mmol) were added and after 5 min additional amine (0.120 mmol) was added. The mixtures were reacted for a further 24 h. The reactions were purified by MDAP (High pH) and concentrated to give:—
Rt
Ex Mass Yield (min)
No. Name Structure (mg) (%) [MH]+ Formic
44 (S)-N3-Methyl-1-(1- phenylethyl)-N5-(2- (tetrahydro-2H-pyran-4- yl)ethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00247
6.8 16 385 0.96
45 (S)-N5-(Cyclopropylmethyl)- N3-methyl-1-(1- phenylethyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00248
6.6 18 327 1.01
46 (S)-N5-Isopropyl-N3- methyl-1-(1-phenylethyl)- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00249
21 61 315 0.97
47 (S)-N5-(4- Hydroxybicyclo[2.2.2]octan- 1-yl)-N3-methyl-1-(1- phenylethyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00250
13 29 397 0.87
48 N5-((1s,4R)-4- Hydroxycyclohexyl)-N3- methyl-1-((S)-1- phenylethyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00251
8 19 371 0.87
49 N5-((1s,4R)-4-Hydroxy-4- methylcyclohexyl)-N3- methyl-1-((S)-1- phenylethyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00252
19 44 385 0.94
Examples 50-64 were prepared as part of an amide array using the following commercial amines,
Reagent
Mass
Reagent Name Structure MW (g) mmol
Ethanamine
Figure US10966961-20210406-C00253
45 0.005 0.120
Propan-1-amine
Figure US10966961-20210406-C00254
59 0.007 0.120
(+/−)-4-Aminobutan-2-ol
Figure US10966961-20210406-C00255
89 0.011 0.120
Tetrahydro-2H-pyran-4-amine
Figure US10966961-20210406-C00256
101 0.012 0.120
(+/−)-Tetrahydrofuran-3-amine
Figure US10966961-20210406-C00257
87 0.010 0.120
(+/−)-2-(Tetrahydro- 2H-pyran-2- yl)ethanamine
Figure US10966961-20210406-C00258
129 0.016 0.120
(+/−)-(Tetrahydrofuran-3- yl)methanamine hydrochloride
Figure US10966961-20210406-C00259
137 0.017 0.120
(+/−)-2-(Tetrahydro- 2H-pyran-3- yl)ethanamine
Figure US10966961-20210406-C00260
129 0.016 0.120
Oxetan-3-ylmethanamine
Figure US10966961-20210406-C00261
87 0.010 0.120
3-Fluorocyclobutanamine, mixture of diastereomers
Figure US10966961-20210406-C00262
89 0.011 0.120
tert-Butyl ((1r,4r)-4-(2- aminoethyl)cyclohexyl) carbamate
Figure US10966961-20210406-C00263
242 0.029 0.120
Oxetan-3-amine
Figure US10966961-20210406-C00264
73 0.009 0.120
2-Cyclopropylethanamine
Figure US10966961-20210406-C00265
85 0.010 0.120
2-(1H-Pyrazol-3-yl)ethanamine
Figure US10966961-20210406-C00266
111 0.013 0.120
(1s,3R,4r,5S,7s)-4- Aminoadamantan-1-ol hydrochloride
Figure US10966961-20210406-C00267
203 0.024 0.120
Method: A stock solution of (5)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (0.027 g, 0.100 mmol)*18=492 mg and HATU (0.038 g, 0.100 mmol)*18=684 mg was dissolved in DMF (0.5 mL)*18=9.0 mL. To this stock solution was added DIPEA (0.052 mL, 0.300 mmol)×18. The vials were capped and shaken at rt to allow full dissolution. After 2 min an aliquot (0.5 mL) of the above stock solution was added to a set of pre-weighed amines*18 (0.120 mmol, for amounts see table) in 1 mL plastic matrix vials. Each vial was capped and shaken to aid dissolution and left to stand overnight for 18 h at rt. T3P (50% by wt. EtOAc) 120 μL and DIPEA (0.052 mL, 0.300 mmol) was added to each reaction mixture and left for >48 h at rt. The reactions were purified by MDAP (high pH) to give final compounds (for amounts see table)
Boc deprotection of the reaction intermediate, corresponding to the coupling of tert-butyl ((1r,4r)-4-(2-aminoethyl)cyclohexyl)carbamate was carried out in DCM (0.5 mL) using 3M HCl in CPME (0.5 mL). A glass microwave vial was used as the reaction vessel. The reaction was left to stir overnight for 22 h at 35° C. Further DCM (0.5 mL) and 3M HCl in CPME (0.5 mL) were added to the reaction mixture and it was left to stir overnight for 18 h at 35° C. The solvent was then removed to dryness under a stream of nitrogen to afford the HCl salt of the free amine. Further DCM (0.5 mL) and 3M HCl in CPME (0.5 mL) were added to the solid material. The solution was left to stand at rt over the weekend. The solvent was removed to dryness to afford the HCl salt of the free amine (example 60) as shown in the table below.
Rt
Ex Mass Yield (min)
No. Name Structure (mg) (%) [MH]+ Formic
50 (S)-N5-Ethyl-N3-methyl- 1-(1-phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00268
18 54 301 0.90
51 (S)-N3-Methyl-1-(1- phenylethyl)-N5-propyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00269
14 40 315 0.98
52 N5-(3-Hydroxybutyl)- N3-methyl-1-((S)-1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00270
19 49 345 0.82
53 (S)-N3-Methyl-1-(1- phenylethyl)-N5- (tetrahydro-2H-pyran- 4-yl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00271
12 31 357 0.89
54 N3-Methyl-1-((S)-1- phenylethyl)-N5- (tetrahydrofuran-3-yl)- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00272
18 48 343 0.86
55 N3-Methyl-1-((S)-1- phenylethyl)-N5-(2- (tetrahydro-2H-pyran- 2-yl)ethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00273
18 43 385 1.05
56 N3-Methyl-1-((S)-1- phenylethyl)-N5- ((tetrahydrofuran-3- yl)methyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00274
18 44 357 0.87
57 N3-Methyl-1-((S)-1- phenylethyl)-N5-(2- (tetrahydro-2H-pyran- 3-yl)ethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00275
21 49 385 0.99
58 (S)-N3-Methyl-N5- (oxetan-3-ylmethyl)-1- (1-phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00276
17 43 343 0.82
59 (S)-N5-(3- Fluorocyclobutyl)-N3- methyl-1-(1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00277
7 19 345 0.98
60 N5-(2-((1r,4S)-4- Aminocyclohexyl)ethyl)- N3-methyl-1-((S)-1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide hydrochloride
Figure US10966961-20210406-C00278
23 47 398 0.60
61 (S)-N3-Methyl-N5- (oxetan-3-yl)-1-(1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00279
14 38 329 0.83
62 (S)-N5-(2- Cyclopropylethyl)-N3- methyl-1-(1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00280
16 43 341 1.07
63 (S)-N5-(2-(1H-Pyrazol- 3-yl)ethyl)-N3-methyl- 1-(1-phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00281
22 54 367 0.82
64 N5-((1S,3S,5R,7R)-5- Hydroxyadamantan-2- yl)-N3-methyl-1-((S)-1- phenylethyl)-1H- pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00282
19 41 423 0.89
Example 65: (S)—N3,N5-Dimethyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00283
To a suspension of N3,N5-dimethyl-1H-pyrazole-3,5-dicarboxamide (39 mg, 0.214 mmol) in THF (1 mL) was added (R)-1-phenylethanol (40 mg, 0.327 mmol) and triphenylphosphine (90 mg, 0.343 mmol) and the reaction mixture was stirred at rt for 15 min. The reaction mixture was cooled to 0° C. and DIAD (0.067 mL, 0.343 mmol) was added. The vial was then heated in the microwave at 140° C. for 15 min. The reaction mixture was then heated at 140° C. for a further 20 min. Further portions of (R)-1-phenylethanol (40 mg, 0.327 mmol), triphenylphosphine (90 mg, 0.343 mmol) and DIAD (0.067 mL, 0.343 mmol) were added and the reaction mixture heated at 140° C. for 15 min. The solvent was removed in vacuo to give 770 mg of a crude yellow oil. This was purified by chromatography on SiO2 (Biotage SNAP 25 g cartridge, eluting with 0-100% ethyl acetate/cyclohexane) to give 50 mg of a colourless oil. This was further purified by MDAP (high pH). The fractions containing desired product were concentrated in vacuo to give (S)—N3,N5-dimethyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (17 mg, 0.053 mmol, 25% yield) as a white solid.
LCMS (2 min Formic): Rt=0.82 min, [MH]+=287.2.
Example 66: 1-Benzyl-N3-((1r,4r)-4-hydroxycyclohexyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00284
To a solution of 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (150 mg, 0.509 mmol) and DIPEA (0.267 mL, 1.527 mmol) in DMF (1 mL) stirred under nitrogen at rt was added HATU (290 mg, 0.764 mmol) followed by the addition of (1r,4r)-4-aminocyclohexanol (70.4 mg, 0.611 mmol) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction mass was poured into ice water, then extracted with EtOAc (3×20 mL). The combined organic layer was washed with water (2×15 mL), brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to afford the crude product. The crude product was triturated with 50% DCM+diethyl ether (3×1 mL), to give solid product, which was filtered and dried under vaccum for 15 min to give, 1-benzyl-N3-((1r,4r)-4-hydroxycyclohexyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (160 mg, 0.447 mmol, 88% yield) as an off-white solid.
LCMS (4.5 min Method A): Rt=1.61 min, [MH]+=357.1.
Example 67: 1-Benzyl-N3-cyclobutyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00285
To a solution of 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (200 mg, 0.733 mmol) and DIPEA (0.384 mL, 2.199 mmol) in DMF (1 mL) stirred under nitrogen at rt was added HATU (418 mg, 1.099 mmol) followed by the addition of cyclobutanamine (104 mg, 1.466 mmol, commercially available from, for example, Sigma Aldrich) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction was poured into ice water, then extracted with EtOAc (3×20 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to give the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 65% EtOAc in n-hexane. The collected pure fractions were concentrated under vaccum to give 1-benzyl-N3-cyclobutyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide (84 mg, 0.255 mmol, 35% yield) as a white solid.
LCMS (10 min Method D): Rt=4.26 min, [MH]+=313.2.
Example 68: (S*)—N5-Cyclopropyl-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00286
Intermediate 11 was purified by chiral HPLC. Preparative NP-HPLC conditions were as follows:—
Column/dimensions: Chiralpak IC (250×30 mm), 5 μm
Mobile Phase: n-hexane: ethanol (70:30)
Flow: 38 mL/min
Temperature: ambient
Wave length: 249 nm
Run time: 18 min
Solubility: THF+n-hexane+ethanol
Load ability/Inj: 33.0 mg/Inj
Total No of injections: 10 The pure fractions corresponding to peak 1 were concentrated under vaccum to give (S*)—N3-cyclopropyl-N3-methyl-1-(1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (36 mg, 0.105 mmol, 15% yield) as a white solid.
LCMS (4.5 min Method B): Rt=1.89 min, [MH]+=313.2.
Example 69: N5-Cyclopropyl-N3-methyl-1-(3-methylbenzyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00287
To a solution of N5-cyclopropyl-N3-methylpyrazole-3,5-dicarboxamide (300 mg, 1.366 mmol) in DMF (2 mL) stirred under nitrogen at rt was added K2CO3 (566 mg, 4.10 mmol) followed by the addition of 1-(bromomethyl)-3-methylbenzene (0.211 mL, 1.640 mmol, commercially available from, for example, Sigma Aldrich) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction was poured into ice water, then extracted with EtOAc (3×25 mL). The combined organic layer was washed with water (3×15 mL), brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to give the crude product which was a mixture of 2 regio-isomers. These were purified by SFC chiral HPLC. Preparative SFC conditions were as follows:—
Column/dimensions: LuxCellulose-2 (250×30 mm), 5 μm
% CO2: 60.0%
% co-solvent: 40.0% (MeOH)
Total Flow: 90.0 g/min
Back Pressure: 100.0 bar
UV: 211 nm
Stack time: 3.8 min
Load/Inj: 16.0 mg
Solubility: methanol
Total No of injections: 20
Instrument details: Make/Model: Thar SFC-200-005
The pure fractions corresponding to peak 1 were concentrated under vaccum to give N5-cyclopropyl-M-methyl-1-(3-methylbenzyl)-1H-pyrazole-3,5-dicarboxamide (76 mg, 0.238 mmol, 63% yield) as a white solid.
LCMS (4.5 min Method B): Rt=1.90 min, [MH]+=313.2.
Example 70: 1-((1H-Indol-5-yl)methyl)-N3-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00288
To a solution of N5-cyclopropyl-N3-methyl-1-((1-tosyl-1H-indol-5-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (420 mg, 0.666 mmol) in methanol (1 mL) stirred under nitrogen at rt was added NaOH (80 mg, 1.999 mmol) in one charge. The reaction mixture was stirred at 70° C. for 16 h. The reaction mass was cooled to rt, then diluted with water (20 mL), then the aqueous layer was extracted with EtOAc (3×15 mL). The combined organic layer was washed with water (3×10 mL), brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to give the crude product as a mixture of regioisomers. These were purified by SFC, chiral HPLC. Preparative SFC conditions were as follows:—
Column/dimensions: Chiralpak IC (250×30 mm), 5 μm
% CO2: 50.0%
% co-solvent: 50.0% (MeOH)
Total Flow: 60.0 g/min
Back Pressure: 100.0 bar
UV: 215 nm
Stack time: 6.5 min
Load/Inj: 8.0 mg
Solubility: MeOH
Total No of injections: 40 The pure fractions corresponding to peak 1 were concentrated under vaccum to give 1-((1H-indol-5-yl)methyl)-N3-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide (95 mg, 0.272 mmol, 85% yield) as an off-white solid.
LCMS (4.5 min Method B): Rt=1.68 min, [MH]+=338.2.
Example 71: 1-((1H-Indol-4-yl)methyl)-N3-cyclopropyl-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00289
To a solution of N5-cyclopropyl-N3-methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (400 mg, 0.745 mmol) in methanol (1 mL) stirred under nitrogen at rt was added NaOH (29.8 mg, 0.745 mmol). The reaction mixture was stirred at 70° C. for 16 h. The reaction was diluted with water, then extracted with DCM (3×15 mL). The combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vaccum to give a crude mixture of regioisomers (410 mg) which were purified by SFC. Preparative SFC conditions were as follows:—
Column/dimensions: Chiralpak AD-H (250×21 mm), 5 μm
% CO2: 60.0%
% co-solvent: 40.0% (MeOH)
Total Flow: 60.0 g/min
Back Pressure: 100.0 bar
UV: 215 nm
Stack time: 3.6 min
Load/Inj: 2.6 mg
Solubility: Methanol+DCM
Total No of injections: 34
The pure fractions corresponding to peak 1 were concentrated under vaccum to give N5-cyclopropyl-N3-methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (400 mg, 0.745 mmol) as an off-white solid.
LCMS (4.5 min Method B): Rt=1.65 min, [MH]+=338.2.
Example 72: N5-((1R,3S,5S,6r)-3-Hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00290
3-Hydroxybicyclo[3.1.0]hexane-6-carboxylic acid (720 mg, 5.06 mmol) was dissolved in toluene (10 mL) and tert-butanol (10 mL), then Et3N (1.412 mL, 10.13 mmol) and diphenyl phosphorazidate (1.637 mL, 7.60 mmol) were added and the mixture was heated at 80° C. overnight. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL), the solvent dried and evaporated to give a pale yellow gum. This was dissolved in DCM (10 mL) and loaded onto a 50 g silica column, then eluted with 0-100% EtOAc/cyclohexane and the ninhydrin active fractions were evaporated in vacuo to give a mixture of diastereomers of tert-butyl (3-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate which was used crude in the following step. tert-Butyl (3-hydroxybicyclo[3.1.0]hexan-6-yl)carbamate (55 mg, 0.258 mmol) (crude) was dissolved in DCM (10 mL) and TFA (1 mL) was added, then the mixture was stirred for 2 h at rt. The solvent was evaporated in vacuo and the residue was dissolved in fresh DCM (10 mL) and Et3N (0.072 mL, 0.516 mmol), HATU (147 mg, 0.387 mmol) and (S)-3-(methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (70.5 mg, 0.258 mmol) were added, then the mixture stirred for 2 h at rt. The reaction mixture was washed with water (10 mL), dried and evaporated in vacuo to give a pale yellow gum. This was purified by MDAP (high pH) to give N5-((1R,3S,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (16 mg, 0.043 mmol, 17% yield) as a colourless solid.
LCMS (2 min High pH): Rt=0.84 min, [MH]+=369.3.
Example 73: N5-(2-((2r,5S)-5-Amino-1,3-dioxan-2-yl)ethyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00291
To a suspension of N5-(2-((2r,5S)-5-(1,3-dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)ethyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (102 mg, 0.192 mmol) in ethanol (3 mL) was added hydrazine hydrate (90 μL, 1.836 mmol). The reaction was stirred at 40° C. for 4 h. The crude product was filtered, and purified by MDAP (high pH). The fractions containing the desired product were concentrated in vacuo to give N5-(2-((2r,5S)-5-amino-1,3-dioxan-2-yl)ethyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (61 mg, 0.137 mmol, 71% yield).
LCMS (2 min High pH): Rt=0.78 min, [MH]+=402.2.
Example 74: N5-(3-((2r,5S)-5-Amino-1,3-dioxan-2-yl)propyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00292
To a suspension of N5-(3-((2r,5S)-5-(1,3-dioxoisoindolin-2-yl)-1,3-dioxan-2-yl)propyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (81 mg, 0.148 mmol) in ethanol (3 mL) was added hydrazine hydrate (90 μL, 1.836 mmol). The reaction was stirred at 40° C. for 19 h. The crude product was filtered, and purified by MDAP (high pH). The fractions containing desired product were concentrated in vacuo to give N5-(3-((2r,5S)-5-amino-1,3-dioxan-2-yl)propyl)-N3-methyl-1-((5)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (42 mg, 0.091 mmol, 61% yield).
LCMS (2 min High pH): Rt=0.80 min, [MH]+=416.3.
Example 75: N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-((R)-2-hydroxy-1-phenylethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00293
N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-1-((R)-2-((tert-butyldimethylsilyl)oxy)-1-phenylethyl)-N5-methylpyrazole-3,5-dicarboxamide (214 mg, 0.442 mmol) was taken up in 4M hydrochloric acid in dioxane (500 μL, 2.000 mmol) and left to stir for 1.5 h. The sample was dissolved in 1:1 MeOH:DMSO (2×1 mL) and purified by MDAP (high pH). The relevant fractions were combined and concentrated in vacuo to give N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-((R)-2-hydroxy-1-phenylethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (68.4 mg, 0.166 mmol, 38% yield) as the second eluting regioisomer.
LCMS (2 min Formic): Rt=0.71 min, [MH]+=371.2.
Example 76: N5-((1R,5S,6s)-3-Azabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00294
To a part suspension of tert-butyl (1R,5S,6s)-6-(3-(methylcarbamoyl)-1-((S)-1-phenylethyl)-1H-pyrazole-5-carboxamido)-3-azabicyclo[3.1.0]hexane-3-carboxylate (95 mg, 0.209 mmol) in DCM (2 mL) was added TFA (0.5 mL, 6.49 mmol) and the reaction mixture was stirred at rt for 6 h. The reaction mixture was concentrated and loaded onto a 2 g SCX cartridge (pre-conditioned with MeOH). This was eluted with MeOH (30 mL) followed by 2M NH3 in MeOH (30 mL). The ammonia fractions containing product were combined and concentrated to give the title compound (57 mg, 0.145 mmol, 69% yield) as a pale yellow oil.
LCMS (2 min Formic): Rt=0.51 min, [MH]+=354.4
Example 77: N5-((1R,5S,6s)-3-Acetyl-3-azabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00295
N5-((1R,5S,6r)-3-Azabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (49 mg, 0.139 mmol) was stirred in acetic anhydride (300 μL, 3.18 mmol) at rt for 2 h. The reaction was diluted with sat NaHCO3 (aq.) and extracted with EtOAc, the organic phase was washed with water, dried using a hydrophobic frit and concentrated to give the title compound (46 mg, 0.105 mmol, 76% yield) as a colourless oil.
LCMS (2 min Formic): Rt=0.77 min, [MH]+=396.4
Example 78: 1-((1H-Indol-4-yl)methyl)-N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-M-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00296
N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((1-tosyl-1H-indol-4-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (99.7 mg, 0.187 mmol) was taken up in THF (4 mL) and methanol (2 mL). Cesium carbonate (304 mg, 0.934 mmol) was added and the reaction was heated to 73° C. and left to stir overnight. The reaction was concentrated in vacuo. Water (10 mL) was added to the residue, which was then acidified with 2N HCl, and then extracted with ethyl acetate (2×10 mL). The combined organics were dried with Na2SO4, filtered and concentrated in vacuo to yield the title compound (63.8 mg, 0.160 mmol, 85% yield) as a purple solid.
LCMS (2 min Formic): Rt=0.74 min, [MH]+=380.3
Example 79: 1-(1H-Indol-4-yl)methyl)-N5-((1R,5S,6s)-3-azabicyclo[3.1.0]hexan-6-yl)-N5-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00297
tert-Butyl (1R,5S,6s)-6-(1-((1H-indol-4-yl)methyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxamido)-3-azabicyclo[3.1.0]hexane-3-carboxylate (86.5 mg, 0.181 mmol) was taken up in DCM (4 mL). TFA (1 mL, 12.98 mmol) was added and the mixture was left to stir at rt for 2 h. The reaction was concentrated in vacuo. The sample was then loaded in methanol and purified by solid phase extraction using a 1 g SCX cartridge, washing with methanol then eluting with 2M ammonia/methanol. The appropriate fractions were combined and evaporated in vacuo to yield the crude product as a cream solid. The crude product was taken up in 1:2 MeOH:DCM (30 mL) and free flow silica added (1 g). The solvent was removed in vacuo and the silica loaded onto a 10 g ULTRA SNAP cartridge in the minimum of DCM and eluted with 0% (2M NH3 in methanol) in DCM for 2CV then 0-20% (2M NH3 in Methanol)/DCM. The appropriate fractions were combined and concentrated in vacuo to yield the desired product, 1-((1H-indol-4-yl)methyl)-N3-((1R,5S,6s)-3-azabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (29.4 mg, 0.070 mmol, 39% yield) as a green solid.
LCMS (2 min Formic): Rt=0.46 min, [MH]+=379.4
Example 80: 1-(1-(1H-Indol-4-yl)ethyl)-N5-((1r,4r)-4-hydroxycyclohexyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00298
To a solution of N5-((1r,4r)-4-hydroxycyclohexyl)-N3-methyl-1-(1-(1-tosyl-1H-indol-4-yl)ethyl)-1H-pyrazole-3,5-dicarboxamide (330 mg, 0.585 mmol) in methanol (2 mL) and THF (4 mL) was added Cs2CO3 (1546 mg, 4.74 mmol) and the reaction mixture was stirred at 70° C. for 2.5 h. The reaction mixture was concentrated in vacuo and partitioned between ethyl acetate and water, and washed with brine. The layers were separated and the aqueous layer was further extracted with ethyl acetate (2×15 mL). The organic layers were combined, dried with a hydrophobic frit and concentrated in vacuo to give 153 mg of crude residue. The crude product was dissolved in 1:1 DMSO:methanol (2 mL) and was purified by MDAP (Formic). The fractions containing desired product were partitioned between sat. NaHCO3 solution and DCM. The layers were separated and the aqueous layer was further extracted with two further portions of DCM (2×15 mL). The organic layers were combined, dried (hydrophobic frit) and concentrated in vacuo to give the title compound (79 mg, 0.193 mmol, 33% yield).
LCMS (2 min Formic): Rt=0.76 min, [M−H]=408
Example 81: 1-((S*)-1-(1H-Indol-4-yl)ethyl)-N3-((1r,4S)-4-hydroxycyclohexyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (single enantiomer of unknown configuration at methyl centre)
Figure US10966961-20210406-C00299
69 mg of 1-(1-(1H-indol-4-yl)ethyl)-N3-((1r,4r)-4-hydroxycyclohexyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (For a preparation see Example 80) was separated by chiral column chromatography using the following conditions:
The crude sample was dissolved in EtOH (4 mL) with heating.
Injection: 2 mL of the solution was injected onto the column.
Solvent: 20% EtOH (+0.2% isopropylamine)/heptane, flow rate=30 mL/min. Wavelength 215 nm
Column: 30 mm×25 cm Chiralpak AD-H (5 μm)
Total number of injections: 2
The fractions corresponding to the first eluting peak were combined and evaporated to give the desired product (30 mg).
LCMS (2 min Formic): Rt=0.74 min, [M−H]=408
Example 82: 1-(1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)ethyl)-N3-methyl-N3-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00300
To a solution of N3-methyl-N3-((1S,2S)-2-methylcyclopropyl)-1-(1-(1-tosyl-1H-pyrrolo[2,3-b]pyridin-4-yl)ethyl)-1H-pyrazole-3,5-dicarboxamide (300 mg, 0.576 mmol) in methanol (2 mL) and THF (4 mL) was added Cs2CO3 (1502 mg, 4.61 mmol) and the reaction mixture (suspension) was stirred at 70° C. for 1.5 h. The reaction mixture was concentrated in vacuo and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer further extracted with ethyl acetate (2×20 mL). The combined organic layers were dried with a hydrophobic frit and concentrated in vacuo to give 152 mg of a crude residue. This was purified by chromatography on silica gel, eluting with 0-100% ethyl acetate/cyclohexane followed by a gradient of 100% ethyl acetate to 25% EtOH/ethyl acetate. The fractions containing desired product were concentrated in vacuo to give the desired product as a pale yellow solid (74 mg, 28% yield).
LCMS (2 min Formic): Rt=0.65 min, [MH]+=367.1
Example 83: 1-((S*)-1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)ethyl)-N3-methyl-N3-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00301
Example 82 (62 mg) was purified by chiral HPLC. The racemate was dissolved in EtOH (4 mL) with heating. Injection: 2 mL of the solution was injected onto the column 20% EtOH (+0.2% isopropylamine)/heptane(+0.2% isopropylamine), flow rate=30 mL/min, detection wavelength=215 nm, 4. Ref 550, 100, Column 30 mm×25 cm Chiralpak AD-H (5 μm), lot no. ADH13231). Total number of injections=2. Fractions from 10-11 min were bulked and labelled peak 1. Fractions from 14-16 min were bulked labelled peak 2. The bulked pure fractions were concentrated in vacuo and then transferred to weighed flasks.
The fractions corresponding to peak 1 were collected to afford example 83 (23 mg)
LCMS (2 min Formic): Rt=0.67 min, [MH]+=367.2.
Example 84: 1-((1H-Indol-5-yl)methyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N5-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00302
N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((1-tosyl-1H-indol-5-yl)methyl)-1H-pyrazole-3,5-dicarboxamide (135 mg, 0.253 mmol) was suspended in THF (2 mL) and methanol (1 mL). Cesium carbonate (412 mg, 1.265 mmol) was added, and the reaction was left to stir at 70° C. for 1 h. The reaction was concentrated in vacuo. Water (20 mL) was added to the residue and this was extracted with ethyl acetate (2×15 mL). The combined organics were dried with Na2SO4, filtered and concentrated in vacuo to yield the desired product (67.4 mg, 0.169 mmol, 67% yield) as a pale brown solid.
LCMS (2 min High pH): Rt=0.76 min, [MH]+=380.5
Example 85: 1-(1-(1H-Indol-5-yl)ethyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00303
N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-(1-(1-tosyl-1H-indol-5-yl)ethyl)-1H-pyrazole-3,5-dicarboxamide (117 mg, 0.214 mmol) was taken up in THF (2 mL) and methanol (1 mL). Cesium carbonate (348 mg, 1.068 mmol) was added, and the reaction left to stir at 70° C. for 2 h. Water (15 mL) was added to the residue, and this was extracted with ethyl acetate (2×15 mL). The combined organics were dried with Na2SO4, filtered and concentrated in vacuo to yield a colourless solid. The crude product was purified by MDAP (Formic). The MDAP did not collect the product, so the waste was concentrated in vacuo and the residue was purified by MDAP (Formic). The MDAP did not collect the product, so the waste was concentrated in vacuo and the residue was purified by MDAP (Formic). The appropriate fractions were dried under a stream of nitrogen to give 1-(1-(1H-indol-5-yl)ethyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (12.2 mg, 0.028 mmol, 13% yield) as a white solid.
LCMS (2 min Formic): Rt=0.82 min, [M−H]=392.
Example 86: 1-(1-(1H-Indol-4-yl)ethyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N5-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00304
To a solution of N5-((1R,5S)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-(1-(1-tosyl-1H-indol-4-yl)ethyl)-1H-pyrazole-3,5-dicarboxamide (278 mg, 0.508 mmol) in methanol (2 mL) and THF (4 mL) was added Cs2CO3 (1323 mg, 4.06 mmol) and the reaction mixture was stirred at 70° C. for 2.5 h. The reaction mixture was concentrated in vacuo and partitioned between ethyl acetate and water. The organic layer was separated and aqueous layer further extracted with ethyl acetate (2×20 mL). The combined organic layers were dried with a hydrophobic frit and concentrated in vacuo to give 152 mg of crude residue which was purified by MDAP (Formic). The fractions containing desired product were partitioned between saturated NaHCO3 solution and DCM. The organic layer was separated and aqueous layer further extracted with DCM (2×20 mL). Combined organic layers were dried (hydrophobic frit) and concentrated in vacuo to give the desired product (44 mg, 0.101 mmol, 20% yield) as a light brown solid.
LCMS (High pH): Rt=0.80 min, [M−H]=392
Example 87: 1-((S*)-1-(1H-Indol-4-yl)ethyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00305
36 mg of 1-(1-(1H-indol-4-yl)ethyl)-N3-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1 pyrazole-3,5-dicarboxamide (for a preparation see Example 86) was separated by chiral column chromatography using the following conditions:
The crude sample was dissolved in EtOH (4 mL) with heating.
Injection: 2 mL of the solution was injected onto the column.
Solvent: 20% EtOH/heptane, flow rate=30 mL/min. Wavelength 215 nm
Column: 30 mm×25 cm Chiralpak AD-H (5 μm)
Total number of injections: 2
The fractions corresponding to the first eluting peak were combined and evaporated to give the desired product (13 mg).
LCMS (Formic): Rt=0.80 min, [M−H]=392
Example 88: 1-((S)-1-(3-Chlorophenyl)ethyl)-N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00306
N5-((1R,5S,6r)-3-((tert-Butyldimethylsilypoxy)bicyclo[3.1.0]hexan-6-yl)-1-((S)-1-(3-chlorophenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (257 mg, 0.497 mmol) was taken up in 4M hydrochloric acid in dioxane (5 mL, 20.00 mmol) and left to stir at rt for 30 min. The reaction was concentrated in vacuo to yield a brown gum. The crude product was purified by MDAP (high pH). The appropriate fractions were concentrated in vacuo to yield 1-((S)-1-(3-chlorophenyl)ethyl)-N3-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (35 mg, 0.083 mmol, 17% yield) as a yellow solid.
LCMS (2 min Formic): Rt=0.89 min, [MH]+=403.6.
The following example was prepared in a similar manner to Example 88:
LCMS:
(System,
Example Structure and name Intermediate used tRET, MH+)
89
Figure US10966961-20210406-C00307
Figure US10966961-20210406-C00308
Formic, 0.88 min, 383.3
Example 90: 1-((S)-1-(3-Chlorophenyl)ethyl)-N3-((1R,3S,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00309
N5-((1R,5S,6r)-3-((tert-Butyldimethylsilypoxy)bicyclo[3.1.0]hexan-6-yl)-1-((S)-1-(3-chlorophenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (257 mg, 0.497 mmol) was taken up in 4M hydrochloric acid in dioxane (5 mL, 20.00 mmol) and left to stir at rt for 30 min. The reaction was concentrated in vacuo to yield a brown gum. The crude product was purified by MDAP (High pH). The desired fractions were concentrated in vacuo to yield 1-((5)-1-(3-chlorophenyl)ethyl)-N5-((1R,3S,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (52.5 mg, 0.124 mmol, 24.91% yield) as a cream solid.
LCMS (2 min Formic): Rt=0.92 min, [MH]+=403.6.
The following example was prepared in a similar manner to Example 90:
LCMS:
(System,
Example Structure and name Intermediate used tRET, MH+)
91
Figure US10966961-20210406-C00310
Figure US10966961-20210406-C00311
Formic, 0.91 min, 383.3
Example 92: 1-Benzyl-N3-cyclopropyl-N3-methyl-1H-Pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00312
To a solution of 1-benzyl-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (1 g, 3.66 mmol) and DIPEA (3.84 mL, 21.99 mmol) in DMF (1 mL) stirred under nitrogen at rt was added oxalyl chloride (0.642 mL, 7.33 mmol) followed by the addition of cyclopropanamine (0.418 g, 7.33 mmol) in one charge. The reaction mixture was stirred at rt for 16 h. The reaction was poured into ice water, then extracted with EtOAc (3×20 mL), combined organic layer was washed with brine solution, dried over sodium sulphate, filtered and the filtrate was concentrated under vacuum to afford the crude product. The crude product was added to a silica gel 60-120 column and was eluted with 65% EtOAc in n-hexane and collected pure fractions were concentrated under vacuum to get 1-benzyl-N3-cyclopropyl-M-methylpyrazole-3,5-dicarboxamide (901 mg, 2.75 mmol, 75% yield) as an off-white solid. LCMS (5.5 min Method E): Rt=2.47 min, [MH]+=299.3.
Examples 93-138
Examples 93-138 were prepared in an analogous manner to examples described above.
Ex Rt (min),
No. Name Structure [MH]+ Method
93 1-Benzyl-N3-methyl-N5- (tetrahydro-2H-pyran-4-yl)- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00313
343.1 1.72 (Method A)
94 1-Benzyl-N3,N5-dimethyl-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00314
273.1 1.63 (Method A)
95 1-Benzyl-N3-methyl-N5-(2- (piperidin-4-yl)ethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00315
370.2 1.34 (Method A)
96 1-Benzyl-N5-isopropyl-N3- methyl-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00316
301.2 1.91 (Method B)
97 1-Benzyl-N3-methyl-N5-propyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00317
301.2 1.93 (Method B)
98 1-Benzyl-N5-ethyl-N3-methyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00318
287.4 1.77 (Method D)
99 1-Benzyl-N3-methyl-N5- (tetrahydrofuran-3-yl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00319
329.3 1.69 (Method B)
100 N5-Cyclopropyl-1-(4- fluorobenzyl)-N3-methyl-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00320
317.1 1.82 (Method B)
101 N5-Cyclopropyl-1-(2- fluorobenzyl)-N3-methyl-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00321
317.1 1.78 (Method E)
102 1-(4-Cyanobenzyl)-N5- cyclopropyl-N3-methyl-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00322
324.1 1.67 (Method B)
103 N5-Cyclopropyl-1-(3- methoxybenzyl)-N3-methyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00323
329.1 6.28 (Method C)
104 N5-Cyclopropyl-1-(4- methoxybenzyl)-N3-methyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00324
329.2 1.82 (Method B)
105 N5-Cyclopropyl-N3-methyl-1- (4-methylbenzyl)-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00325
313.1 6.63 (Method C)
106 N5-Cyclopropyl-1-(2- methoxybenzyl)-N3-methyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00326
329.2 1.76 (Method B)
107 1-((1H-Indol-4-yl)methyl)-N5- ((1R,5S,6s)-3-acetyl-3- azabicyclo[3.1.0]hexan-6-yl)- N3-methyl-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00327
421.3 0.69 (Formic)
108 N3-Ethyl-N5-((1r,4S)-4- hydroxycyclohexyl)-1-((S)-1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00328
385.3 0.88 (Formic)
109 N5-((1R,5S,6r)-3- Oxabicyclo[3.1.0]hexan-6-yl)- N3-methyl-1-((R)-1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00329
355.3 0.85 (Formic)
110 N5-((1r,4R)-4- Hydroxycyclohexyl)-N3- methyl-1-((R)-1-phenylethyl)- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00330
371.3 0.82 (Formic)
111 N5-(1-Hydroxypropan-2-yl)-N3- methyl-1-((S)-1-phenylethyl)- 1H-pyrazole-3,5- dicarboxamide, mixture of diastereomers
Figure US10966961-20210406-C00331
331.1 0.79 (Formic)
112 (S)-N5-(tert-Butyl)-N3-methyl- 1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00332
329.1 1.08 (Formic)
113 N5-((S)-sec-Butyl)-N3-methyl- 1-((S)-1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00333
329.2 1.04 (Formic)
114 N5-((1R,3R,5S,6s)-3- Hydroxybicyclo[3.1.0]hexan-6- yl)-N3-methyl-1-((S)-1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00334
369.4 0.79 (High pH)
115 (S)-N5-(2-Hydroxyethyl)-N3- methyl-1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00335
317.1 0.75 (Formic)
116 (S)-N5-(2-Cyanoethyl)-N3- methyl-1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00336
326.1 0.86 (Formic)
117 (S)-N5-(2-(1H-Imidazol-4- yl)ethyl)-N3-methyl-1-(1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00337
367.1 0.53 (Formic)
118 (S)-N3-Methyl-N5-(3- morpholinopropyl)-1-(1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00338
400.2 0.54 (Formic)
119 (S)-N3-Methyl-1-(1- phenylethyl)-N5-(2-(pyridin-4- yl)ethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00339
378.1 0.56 (Formic)
120 (S)-N5-(3- (Dimethylamino)propyl)-N3- methyl-1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00340
358.1 0.53 (Formic)
121 (S)-N5-(2-Methoxyethyl)-N3- methyl-1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00341
331.1 0.87 (Formic)
122 (S)-N5-(2-(1H-Imidazol-2- yl)ethyl)-N3-methyl-1-(1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00342
367.1 0.52 (Formic)
123 (S)-N3-Methyl-1-(1- phenylethyl)-N5-(pyridin-4- ylmethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00343
364.1 0.56 (Formic)
124 (S)-N3-Methyl-1-(1- phenylethyl)-N5-(2-(pyridin-3- yl)ethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00344
378.1 0.58 (Formic)
125 (S)-N3-Methyl-1-(1- phenylethyl)-N5-(2-(pyridin-2- yl)ethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00345
378.1 0.59 (Formic)
126 (S)-N5-(2,2-Difluoroethyl)-N3- methyl-1-(1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00346
337.1 0.95 (Formic)
127 N5-(2-(4-Acetylmorpholin-2- yl)ethyl)-N3-methyl-1-((S)-1- phenylethyl)-1H-pyrazole-3,5- dicarboxamide, mixture of diastereomers
Figure US10966961-20210406-C00347
428.2 0.81 (Formic)
128 N5-((1s,3R)-3- Hydroxycyclobutyl)-N3-methyl 1-((S)-1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00348
343.1 0.80 (Formic)
129 N3-Methyl-1-((S)-1- phenylethyl)-N5-((tetrahydro- 2H-pyran-2-yl)methyl)-1H- pyrazole-3,5-dicarboxamide, mixture of diastereomers
Figure US10966961-20210406-C00349
371.1 1.01 (Formic)
130 N5-((1R,2R)-2- Hydroxycyclobutyl)-N3-methyl- 1-((S)-1-phenylethyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00350
343.1 0.82 (Formic)
131 1-((R*)-1-(1H-Indol-4- yl)ethyl)-N5-(1r,4R)-4- hydroxycyclohexyl)-N3-methyl- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00351
MH− 408.3 0.74 (Formic)
132 1-((R)-1-(1H-Indol-4- yl)ethyl)-N5-((1R,5S,6r)-3- oxabicyclo[3.1.0]hexan-6-yl)- N3-methyl-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00352
MH− 392.3 0.77 (Formic)
133 N5-((1R,5S,6r)-3- Oxabicyclo[3.1.0]hexan-6-yl)- N3-methyl-1-((S)-1-(pyridin-2- yl)ethyl)-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00353
356.5 0.67 (High pH)
134 (S)-N3-Methyl-N5-(1H-pyrazol- 4-yl)-1-(1-(pyridin-2-yl)ethyl)- 1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00354
340.5 0.53 (Formic)
135 N3-Methyl-N5-((1S,2S)-2- methylcyclopropyl)-1-(1-(1- tosyl-1H-pyrrolo[2,3-b]pyridin- 4-yl)ethyl)-1H-pyrazole-3,5- dicarboxamide, mixture of diasatereomers
Figure US10966961-20210406-C00355
521.1 1.11 (Formic)
136 N5-((1R,5S,6r)-3- Oxabicyclo[3.1.0]hexan-6-yl)- 1-((S)-1-(2- methoxyphenyl)ethyl)-N3- methyl-1H-pyrazole-3,5- dicarboxamide
Figure US10966961-20210406-C00356
385.2 0.84 (Formic)
137 1-((R)-1-(1H-Pyrrolo[2,3- b]pyridin-4-yl)ethyl)-N3- methyl-N5-((1S,2S)-2- methylcyclopropyl)-1H- pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00357
367.2 0.66 (Formic)
138 1-((S)-1-(3- Chlorophenyl)ethyl)-N5- ((1R,3R,5S,6s)-3- hydroxybicyclo[3.1.0]hexan-6- yl)-N3-methyl-1H-pyrazole- 3,5-dicarboxamide
Figure US10966961-20210406-C00358
403.6 0.87 (Formic)
Example 139: N5-((1r,4.5)-4-Methoxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00359
(S)-3-(Methylcarbamoyl)-1-(1-phenylethyl)-1H-pyrazole-5-carboxylic acid (For a preparation, see Intermediate 5, 112 mg, 0.410 mmol) was dissolved in DMF (1 mL) before the addition of HATU (234 mg, 0.615 mmol). (1r,4r)-4-Methoxycyclohexan-1-amine hydrochloride (82 mg, 0.445 mmol) was then added followed by DIPEA (0.358 mL, 2.049 mmol) and the reaction stirred at rt for 2 h. The sample was purified directly by MDAP (high pH). The relevant fractions were combined and concentrated in vacuo to give N5-((1r,4S)-4-methoxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide (135 mg, 0.316 mmol, 77% yield).
LCMS (2 min Formic): Rt=0.96 min, [MH]+=385.2.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.32 (d, J=7.83 Hz, 1H) 8.09-8.17 (m, 1H) 7.20-7.34 (m, 6H) 6.67 (q, J=7.01 Hz, 1H) 3.60-3.74 (m, 1H) 3.23 (s, 3H) 3.03-3.14 (m, 1H) 2.78 (d, J=4.65 Hz, 3H) 1.94-2.05 (m, 2H) 1.84 (d, J=7.09 Hz, 4H) 1.71-1.79 (m, 1H) 1.13-1.40 (m, 4H)
Example 140: N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-1-(indolin-4-ylmethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00360
1-((1H-Indol-4-yl)methyl-N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H pyrazole-3,5-dicarboxamide (For a preparation, see Example 78, 10 mg, 0.026 mmol) was taken up in glacial acetic acid (1 mL). Sodium cyanoborohydride (3.31 mg, 0.053 mmol) was added, and the reaction left to stir at rt for 2 h. The reaction was diluted with water and extracted with 10% MeOH in DCM. The aqueous layer was concentrated in vacuo and extracted with 10% MeOH in DCM. The combined organics were passed through a hydrophobic frit and concentrated in vacuo to give a crude residue which was purified by MDAP (High pH). The appropriate fractions were concentrated in vacuo to give N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-(indolin-4-ylmethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (3.8 mg, 8.47 μmol, 32% yield) as a white solid.
LCMS (2 min HpH): Rt=0.69 min, [MH]+=382.2
Example 141: 1-(1-(3-(2-Hydroxyethoxy)phenyl)ethyl)-N3-methyl-N3-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide, Mixture of Diastereomers
Figure US10966961-20210406-C00361
1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-N3-methyl-N5-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide (For a preparation, see Intermediate 132, 1.1 g, 2.197 mmol, mixture of diastereomers) was dissolved in DCM (20 mL) and HCl (5 mL, 5.00 mmol, 1M in ether) was added, then the mixture was stirred at rt for 2 h. The solvent was evaporated in vacuo and the residue was dissolved in DCM (10 mL) and loaded onto a 25 g SNAP Ultra silica column, which was eluted with 0-100% (25% EtOH/EtOAc)/cyclohexane and the product-containing fractions were evaporated in vacuo to give 1-(1-(3-(2-hydroxyethoxy)phenyl)ethyl)-N3-methyl-N5-((1S,2S)-2-methylcyclopropyl)-1H-pyrazole-3,5-dicarboxamide (0.71 g, 1.837 mmol, 84% yield, mixture of diastereomers) as a colourless solid.
LCMS (2 min Formic): Rt=0.81 min, [M+Na]+=409.3.
Example 142: (+/−)-N5-((1R,5S,6r)-3-Oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(3-(2-hydroxyethoxy)phenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide
Figure US10966961-20210406-C00362
(+/−)-1-(1-(3-(2-((tert-Butyldimethylsilypoxy)ethoxy)phenyl)ethyl)-3-(methylcarbamoyl)-1H-pyrazole-5-carboxylic acid (For a preparation, see Intermediate 131, 0.27 g, 0.603 mmol), (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-amine, hydrochloride (0.106 g, 0.784 mmol), HATU (0.298 g, 0.784 mmol) and Et3N (0.252 mL, 1.810 mmol) were dissolved in DCM (20 mL) and the mixture was stirred for 2 h, then allowed to stand over the weekend at rt. The resulting mixture was stirred with water (50 mL) for 1 h, then the organic layer was separated, dried and evaporated in vacuo to give a pale yellow gum. This was dissolved in DCM and loaded onto a 100 g SNAP ultra silica column and eluted with 0-100% EtOAc/cyclohexane. The product-containing fractions were evaporated in vacuo to give (+/−)-N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)phenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (0.14 g, 0.265 mmol, 44% yield) as a colourless solid. This solid was dissolved in DCM (5 mL) and HCl (2 mL, 2.0 mmol, 1M in ether) was added, then the mixture was stirred for 2 h at rt. The solvent was evaporated in vacuo and the crude product was dissolved in DCM and loaded onto a 10 g silica column and purified by flash chromatography eluting with 0-100% (25% EtOH/EtOAc)/cyclohexane. The product-containing fractions were evaporated in vacuo to give (+/−)-N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(3-(2-hydroxyethoxy)phenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide (81 mg, 0.195 mmol, 32% yield) as a colourless foam.
LCMS (2 min Formic): Rt=0.71 min, [MH]+=415.4.
The following examples were prepared in a similar manner to Example 1:
Example
number Structure and name [MH+] Rt (min)
143
Figure US10966961-20210406-C00363
370.0 1.00 (Formic)
(S)-N3-Methyl-N5-(3-methylisothiazol-5-
yl)-1-(1-phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
144
Figure US10966961-20210406-C00364
493.1 1.30 (High pH)
N5-((1R,3R,5S,6r)-3-
Methoxybicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
145
Figure US10966961-20210406-C00365
383.4 0.86 (Formic)
(S)-N5-(6-Hydroxyspiro[3.3]heptan-2-
yl)-N3-methyl-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide, mixture of
diastereomers
146
Figure US10966961-20210406-C00366
385.3 0.89 (High pH)
N5-((1r,4S)-4-
(Hydroxymethyl)cyclohexyl)-N3-methyl-
1-((S)-1-phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
147
Figure US10966961-20210406-C00367
351.0 0.81 (Formic)
(S)-N3-Methyl-1-(1-phenylethyl)-N5-
(pyridazin-4-yl)-1H-pyrazole-3,5-
dicarboxamide
148
Figure US10966961-20210406-C00368
399.1 0.94 (High pH)
N5-((1S*,4S*)-4-Hydroxy-3,3-
dimethylcyclohexyl)-N3-methyl-1-((S)-
1-phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
149
Figure US10966961-20210406-C00369
369.5 0.94 (High pH)
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S)-1-(m-tolyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
150
Figure US10966961-20210406-C00370
407.2 1.00 (Formic)
(+/−)-N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(3-
chloro-5-fluorophenyl)ethyl)-N3-methyl-
1H-pyrazole-3,5-dicarboxamide
151
Figure US10966961-20210406-C00371
399.2 1.01 (High pH)
N5-((1s*,4R*)-4-Ethyl-4-
hydroxycyclohexyl)-N3-methyl-1-((S)-
1-phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
152
Figure US10966961-20210406-C00372
343.0 0.75 (Formic)
N5-((1r*,3S*)-3-Hydroxycyclobutyl)-N3-
methyl-1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
153
Figure US10966961-20210406-C00373
366.0 0.75 (Formic)
(S)-N3-Methyl-N5-(6-oxo-1,6-
dihydropyridin-3-yl)-1-(1-phenylethyl)-
1H-pyrazole-3,5-dicarboxamide
154
Figure US10966961-20210406-C00374
419.3 0.98 (High pH)
1-((S)-1-(3-Chlorophenyl)ethyl)-N5-
((1r,4S)-4-(hydroxymethyl)cyclohexyl)-
N3-methyl-1H-pyrazole-3,5-
dicarboxamide
155
Figure US10966961-20210406-C00375
419.3 0.98 (Formic)
1-((S)-1-(3-Chlorophenyl)ethyl)-N5-
((1S,3S,4S)-4-hydroxy-3-
methylcyclohexyl)-N3-methyl-1H-
pyrazole-3,5-dicarboxamide
156
Figure US10966961-20210406-C00376
357.2 0.91 (High pH)
(+/−)-N5-(1r,3S)-3-
Methoxycyclobutyl)-N3-methyl-1-(1-
phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
157
Figure US10966961-20210406-C00377
377.1 0.87 (High pH)
1-((S)-1-(3-Chlorophenyl)ethyl)-N5-
((trans)-2-
(hydroxymethyl)cyclopropyl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide,
mixture of diastereomers
158
Figure US10966961-20210406-C00378
391.3 0.89 (Formic)
1-((S)-1-(3-Chlorophenyl)ethyl)-N5-
((trans)-3-hydroxycyclopentyl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide,
mixture of diastereomers
159
Figure US10966961-20210406-C00379
387.3 0.93 (Formic)
(+/−)-N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(3-
fluoro-2-methylphenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide
160
Figure US10966961-20210406-C00380
387.3 0.93 (Formic)
N5-((1s,4R)-4-Hydroxycyclohexyl)-N3-
methyl-1-((S*)-1-(3-
(trifluoromethyl)phenyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
161
Figure US10966961-20210406-C00381
341.2 0.80 (Formic)
1-Benzyl-N5-((1R,5S,6r)-3-
oxabicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide
162
Figure US10966961-20210406-C00382
493.5 1.30 (High pH)
N5-((1R*,3S*,5S*,6s*)-3-
(Benzyloxy)bicyclo[3.1.0]hexan-6-yl)-1-
((S)-1-(3-chlorophenyl)ethyl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide
163
Figure US10966961-20210406-C00383
417.4 0.97 (Formic)
(S)-1-(1-(3-Chlorophenyl)ethyl)-N5-(6-
hydroxyspiro[3.3]heptan-2-yl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide,
mixture of diastereomers
164
Figure US10966961-20210406-C00384
385.3 0.86 (Formic)
N5-((1S,3S,4S)-4-Hydroxy-3-
methylcyclohexyl)-N3-methyl-1-((S)-1-
phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
165
Figure US10966961-20210406-C00385
368.1 1.00 (Formic)
(S)-N5-(2-Ethyl-2H-1,2,3-triazol-4-yl)-
N3-methyl-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
166
Figure US10966961-20210406-C00386
399.2 0.91 (HpH)
N5-((1r*,4S*)-4-Ethyl-4-
hydroxycyclohexyl)-N3-methyl-1-((S)-1-
phenylethyl)-1H-pyrazole-3,5-
dicarboxamide
167
Figure US10966961-20210406-C00387
437.2 0.92 (Formic)
N5-((1R*,3R*,5S*,6r*)-3-
Hydroxybicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S*)-1-(3-
(trifluoromethyl)phenyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
168
Figure US10966961-20210406-C00388
399.6 0.95 (HpH)
N5-((1S,3S,4S)-4-Hydroxy-3-
methylcyclohexyl)-N3-methyl-1-((S)-1-
(m-tolyl)ethyl)-1H-pyrazole-3,5-
dicarboxamide
169
Figure US10966961-20210406-C00389
403.6 0.93 (HpH)
1-((S)-1-(3-Fluorophenyl)ethyl)-N5-
((1S,3S,4S)-4-hydroxy-3-
methylcyclohexyl)-N3-methyl-1H-
pyrazole-3,5-dicarboxamide
170
Figure US10966961-20210406-C00390
439.1 0.96 (Formic)
(+/−)-N5-((1r,4r)-4-
Hydroxycyclohexyl)-N3-methyl-1-(1-(4-
(trifluoromethyl)phenyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
171
Figure US10966961-20210406-C00391
343.1 0.78 (HpH)
N5-((1S*,2S*)-2-
(Hydroxymethyl)cyclopropyl)-N3-
methyl-1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
172
Figure US10966961-20210406-C00392
364.0 0.75 (Formic)
(S)-N3-Methyl-N5-(5-methylpyridin-3-
yl)-1-(1-phenylethyl)-1H-pyrazole-3,5-
dicarboxamide, formate salt
173
Figure US10966961-20210406-C00393
350.0 0.75 (Formic)
(S)-N3-Methyl-1-(1-phenylethyl)-N5-
(pyridin-3-yl)-1H-pyrazole-3,5-
dicarboxamide
174
Figure US10966961-20210406-C00394
369.3 0.90 (Formic)
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S)-1-(o-tolyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
175
Figure US10966961-20210406-C00395
377.2 0.87 (High pH)
1-((S)-1-(3-Chlorophenyl)ethyl)-N5-
((1r,3S)-3-hydroxycyclobutyl)-N3-
methyl-1H-pyrazole-3,5-dicarboxamide
176
Figure US10966961-20210406-C00396
437.2 0.95 (Formic)
N5-((1R*,3S*,5S*,6r*)-3-
Hydroxybicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S*)-1-(3-
(trifluoromethyl)phenyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
177
Figure US10966961-20210406-C00397
369.6 0.95 (High pH)
N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-N3-
methyl-1-((S)-1-(p-tolyl)ethyl)-1H-
pyrazole-3,5-dicarboxamide
178
Figure US10966961-20210406-C00398
350.0 0.98 (Formic)
(S)-N3-Methyl-1-(1-phenylethyl)-N5-
(pyridin-2-yl)-1H-pyrazole-3,5-
dicarboxamide formate salt
179
Figure US10966961-20210406-C00399
371.6 0.98 (High pH)
N5-((1S,3S)-3-Methoxycyclopentyl)-N3-
methyl-1-((S)-1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
180
Figure US10966961-20210406-C00400
407.3 0.98 (Formic)
(+/−)-N5-((1R,5S,6r)-3-
Oxabicyclo[3.1.0]hexan-6-yl)-1-(1-(4-
chloro-3-fluorophenyl)ethyl)-N3-methyl-
1H-pyrazole-3,5-dicarboxamide
181
Figure US10966961-20210406-C00401
335.2 1.00 (Formic)
(S)-1-(1-(3-Chlorophenyl)ethyl)-N5-
ethyl-N3-methyl-1H-pyrazole-3,5-
dicarboxamide
182
Figure US10966961-20210406-C00402
382.0 1.06 (Formic)
(S)-N5-(3-Ethyl-4-methylisoxazol-5-yl)-
N3-methyl-1-(1-phenylethyl)-1H-
pyrazole-3,5-dicarboxamide
183
Figure US10966961-20210406-C00403
356.0 1.00 (Formic)
(S)-N3-Methyl-1-(1-phenylethyl)-N5-
(thiazol-2-yl)-1H-pyrazole-3,5-
dicarboxamide
The following examples were prepared in a similar manner to Example 81:
Example
number Structure and name [MH+] Rt (min)
184
Figure US10966961-20210406-C00404
403.5 0.94 (High pH)
185
Figure US10966961-20210406-C00405
391.3 0.90 (Formic)
186
Figure US10966961-20210406-C00406
357.4 0.80 (Formic)
187
Figure US10966961-20210406-C00407
391.3 0.89 (Formic)
188
Figure US10966961-20210406-C00408
387.2 0.95 (Formic)
189
Figure US10966961-20210406-C00409
439.3 0.95 (Formic)
190
Figure US10966961-20210406-C00410
387.2 0.92 (Formic)
191
Figure US10966961-20210406-C00411
405.5 0.83 (High pH)
192
Figure US10966961-20210406-C00412
371.6 0.97 (High pH)
193
Figure US10966961-20210406-C00413
439.3 0.95 (Formic)
194
Figure US10966961-20210406-C00414
357.4 0.79 (Formic)
195
Figure US10966961-20210406-C00415
417.3 1.01 (Formic)
196
Figure US10966961-20210406-C00416
394.2 [MH+] 0.80 (Formic)
197
Figure US10966961-20210406-C00417
437.3 [MH+] 0.94 (Formic)
198
Figure US10966961-20210406-C00418
387.1 [MH+] 0.95 (Formic)
199
Figure US10966961-20210406-C00419
371.6 [MH+] 0.97 (HpH)
200
Figure US10966961-20210406-C00420
417.3 1.01 (Formic)
201
Figure US10966961-20210406-C00421
403.5 0.94 (High pH)
202
Figure US10966961-20210406-C00422
383.3 0.85 (Formic)
203
Figure US10966961-20210406-C00423
343.3 0.78 (High pH)
204
Figure US10966961-20210406-C00424
407.3 1.00 (Formic)
205
Figure US10966961-20210406-C00425
383.3 0.85 (Formic)
206
Figure US10966961-20210406-C00426
343.2 0.78 (High pH)
The following examples were prepared in a similar manner to Example 88:
Example
number Name and structure [MH+] Rt (min)
207
Figure US10966961-20210406-C00427
387.6 0.89 (high pH)
208
Figure US10966961-20210406-C00428
383.6 0.92 (High pH)
209
Figure US10966961-20210406-C00429
387.6 0.86 (High pH)
210
Figure US10966961-20210406-C00430
437.2 0.99 (Formic)
211
Figure US10966961-20210406-C00431
383.5 0.89 (HpH)
212
Figure US10966961-20210406-C00432
377.1 0.87 (HpH)
The following example was prepared in a similar manner to Example 74:
Example
number Structure and name [MH+] Rt (min)
213
Figure US10966961-20210406-C00433
450.6 0.91 (High pH)
The following examples were prepared in a similar manner to Example 75:
Example
number Structure and name [MH+] Rt (min)
214
Figure US10966961-20210406-C00434
407.2 0.85 (High pH)
215
Figure US10966961-20210406-C00435
405.5 0.84 (High pH)
216
Figure US10966961-20210406-C00436
435.2 0.90 (High pH)
217
Figure US10966961-20210406-C00437
401.4 0.79 (Formic)
The following examples were prepared in a similar manner to Example 86:
Example
number Structure and Name [MH+] Rt (min)
218
Figure US10966961-20210406-C00438
380.3 0.89 (Formic)
219
Figure US10966961-20210406-C00439
381.3 0.66 (Formic)
Examples 220-261 were prepared in an analogous manner to examples described above.
Example
number Structure and name [MH+] Rt (min)
220
Figure US10966961-20210406-C00440
416.2 [M + Na] 0.80 (Formic)
221
Figure US10966961-20210406-C00441
407.3 0.98 (Formic)
222
Figure US10966961-20210406-C00442
399.1 1.01 (High pH)
223
Figure US10966961-20210406-C00443
357.0 0.90 (Formic)
224
Figure US10966961-20210406-C00444
273.2 0.77 (Formic)
225
Figure US10966961-20210406-C00445
365.4 0.67 (High pH)
226
Figure US10966961-20210406-C00446
383.6 0.98 (High pH)
227
Figure US10966961-20210406-C00447
392.5 0.80 (Formic)
228
Figure US10966961-20210406-C00448
378.3 [M − H] 0.80 (High pH)
229
Figure US10966961-20210406-C00449
437.2 0.94 (Formic)
230
Figure US10966961-20210406-C00450
403.6 0.97 (High pH)
231
Figure US10966961-20210406-C00451
385.3 0.91 (Formic)
232
Figure US10966961-20210406-C00452
399.6 1.00 (High pH)
233
Figure US10966961-20210406-C00453
338.2 1.70 (Formic)
234
Figure US10966961-20210406-C00454
338.2 1.67 (Formic)
235
Figure US10966961-20210406-C00455
419.3/420.4 1.02 (Formic)
236
Figure US10966961-20210406-C00456
399.6 1.02 (High pH)
237
Figure US10966961-20210406-C00457
407.3 0.98 (Formic)
238
Figure US10966961-20210406-C00458
437.2 0.96 (Formic)
239
Figure US10966961-20210406-C00459
379.0 1.00 (Formic)
240
Figure US10966961-20210406-C00460
425.0 1.20 (Formic)
241
Figure US10966961-20210406-C00461
437.2 0.93 (Formic)
242
Figure US10966961-20210406-C00462
407.3 1.00 (Formic)
243
Figure US10966961-20210406-C00463
378.1 0.62 (Formic)
244
Figure US10966961-20210406-C00464
329.2 1.84 (Formic)
245
Figure US10966961-20210406-C00465
313.1 6.68 (TFA 15 min method)
246
Figure US10966961-20210406-C00466
409.0 1.05 (Formic)
247
Figure US10966961-20210406-C00467
371.3 0.68 (Formic)
248
Figure US10966961-20210406-C00468
368.0 0.95 (Formic)
249
Figure US10966961-20210406-C00469
439.3 0.95 (Formic)
250
Figure US10966961-20210406-C00470
439.3 0.95 (Formic)
251
Figure US10966961-20210406-C00471
405.5 0.83 (High pH)
252
Figure US10966961-20210406-C00472
317.2 1.86 (Formic)
253
Figure US10966961-20210406-C00473
329.2 1.78 (Formic)
254
Figure US10966961-20210406-C00474
387.2 0.92 (Formic)
255
Figure US10966961-20210406-C00475
317.1 1.80 (Formic)
256
Figure US10966961-20210406-C00476
313.2 1.92 (Formic)
257
Figure US10966961-20210406-C00477
339.2 1.93 (Formic)
258
Figure US10966961-20210406-C00478
396.2 0.92 (High pH)
259
Figure US10966961-20210406-C00479
395.3 3.83 (Formic 10 min method)
260
Figure US10966961-20210406-C00480
324.1 1.71 (Formic)
261
Figure US10966961-20210406-C00481
329.1 6.35 (TFA 15 min method)

Biological Data
The compounds of formula (I) may be tested in one or more of the following assays:
Time Resolved Fluorescence Resonance Energy Transfer (TR-FRET) assay
Bromodomain binding was assessed utilising a time resolved fluorescent resonance energy transfer (TR-FRET) competition assay. To enable this approach a known, high affinity, pan-BET interacting small molecule was labelled with Alexa Fluor® 647, which is a far-red-fluorescent dye (Reference Compound X). Reference Compound X acts as a reporter of bromodomain binding and is the acceptor fluorophore component of the TR-FRET pair. Europium chelate, conjugated to an anti-6*His antibody, was utilised as the donor fluorophore in the TR-FRET pair. The anti-6*His antibody binds selectively to a six Histidine purification epitope added to the amino-terminus of each of the BET tandem bromodomain protein constructs used in this study. A TR-FRET signal is generated when the donor and acceptor fluorophores are in close proximity, between 20-80 Å, which is enabled in this assay by binding of Reference Compound X to the bromodomain protein.
Reference Compound X: 4-((Z)-3-(6-((5-(2-((4.5)-6-(4-chlorophenyl)-8-methoxy-1-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)acetamido)pentyl)amino)-6-oxohexyl)-2-((2E,4E)-5-(3,3-dimethyl-5-sulfo-1-(4-sulfobutyl)-3H-indol-1-ium-2-yl)penta-2,4-dien-1-ylidene)-3-methyl-5-sulfoindolin-1-yl)butane-1-sulphonate)
Figure US10966961-20210406-C00482
To a solution of N-(5-aminopentyl)-2-((45)-6-(4-chlorophenyl)-8-methoxy-1-methyl-4H-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)acetamide (for a preparation see Reference Compound J, WO2011/054848A1, 1.7 mg, 3.53 μmol) in DMF (40 μL) was added a solution of AlexaFluor647-ONSu (2.16 mg, 1.966 μmol) also in DMF (100 μL). The mixture was basified with DIPEA (1 μl, 5.73 μmol) and agitated overnight on a vortex mixer.
The reaction mixture was evaporated to dryness. The solid was dissolved in MeCN/water/AcOH (5/4/1, <1 mL) filtered and was applied to a Phenomenex Jupiter C18 preparative column and eluted with the following gradient (A=0.1% trifluoroacetic acid in water, B=0.1% TFA/90% MeCN/10% water): Flow rate=10 mL/min., AU=20/10 (214 nm):
5-35%, t=0 min: B=5%; t=10 min: B=5%; t=100 min: B=35%; t=115 min: B=100% (Sep. grad: 0.33%/min)
The major component was eluted over the range 26-28% B but appeared to be composed of two peaks. The middle fraction (F1.26) which should contain “both” components was analysed by analytical HPLC (Spherisorb ODS2, 1 to 35% over 60 min): single component eluting at 28% B.
Fractions F1.25/26&27 were combined and evaporated to dryness. Transferred with DMF, evaporated to dryness, triturated with dry ether and the blue solid dried overnight at <0.2 mbar: 1.54 mg.
Analytical HPLC (Sphersisorb ODS2, 1 to 35% B over 60 min): MSM10520-1: [M+H]+ (obs): 661.8/− corresponding with N5-29. This equates to [(M+2H)/2]+ for a calculated mass of 1320.984 which is N5-29. This is a standard occurrence with the Alexa Fluor 647 dye and represents a theoretical loss of two methylene groups under the conditions of the mass spectrometer.
Assay Principle: In order to generate a TR-FRET signal, donor fluorophore is excited by a laser at A337 nm, which subsequently leads to emission at λ618 nm. If the acceptor fluorophore is in close proximity then energy transfer can occur, which leads to emission of Alexa Fluor® 647 at λ665 nm. In the presence of competitor compound, Reference Compound X can be displaced from binding to the bromodomain. If displacement occurs, the acceptor fluorophore is no longer in proximity to the donor fluorophore, which prevents fluorescent energy transfer and, subsequently, a loss of Alexa Fluor® 647 emission at λ665 nm.
The competition of the compounds of formula (I) with Reference Compound X for binding to the BET family (BRD2, BRD3, BRD4 and BRDT) was assessed using protein truncates spanning both bromodomain 1 (BD1) and bromodomain 2 (BD2). In order to monitor differential binding to either BD1 or BD2, single residue mutations of key tyrosines to alanine were made in the acetyl lysine binding pockets. To validate this approach, a double residue mutant tandem domain protein was produced for each of the BET family members. Utilising a Fluorescence Polarisation approach, binding affinities for each of the single and double mutants for Reference Compound X were determined. The affinities of the double mutant tandem proteins for Reference Compound X were greatly greatly reduced in comparison to the non mutated, wild type tandem BET proteins (>1000 fold reduction in Kd). The affinities of the single mutated bromdomain tandem proteins for Reference Compound X were equi-potent with the corresponding non-mutated BET protein. These data demonstrated that single mutations of Tyrosine to Alanine reduce the Kd of the interaction between the mutated bromodomain and Reference Compound X by >1000 fold. In the TR-FRET competition assay, Reference Compound X is used at a concentration that is equivalent to the Kd for the non-mutated bromodomain, which ensures that no binding at the mutated bromodomain is detected.
Protein production: Recombinant Human Bromodomains [(BRD2 (1-473) (Y113A) and (Y386A), BRD3 (1-435) (Y73A) and (Y348A) BRD4 (1-477) (Y97A) and (Y390A) and BRDT (1-397) (Y66A) and (Y309A)] were expressed in E. coli cells (in pET15b vector for BRD2/3/4 and in pET28a vector for BRDT) with a 6-His tag at the N-terminal. The His-tagged Bromodomain pellet was resuspended in 50 mM HEPES (pH7.5), 300 mM NaCl, 10 mM imidazole & 1 μL/mL protease inhibitor cocktail and extracted from the E. coli cells using sonication and purified using a nickel sepharose high performance column, the proteins were washed and then eluted with a linear gradient of 0-500 mM imidazole with buffer 50 mM HEPES (pH7.5), 150 mM NaCl, 500 mM imidazole, over 20 column volumes. Final purification was completed by Superdex 200 prep grade size exclusion column. Purified protein was stored at −80° C. in 20 mM HEPES pH 7.5 and 100 mM NaCl. Protein identity was confirmed by peptide mass fingerprinting and predicted molecular weight confirmed by mass spectrometry. Protocol for Bromodomain BRD2, 3, 4 and T, BD1+BD2 mutant TR-FRET competition assays:
All assay components were dissolved in an assay buffer composing of 50 mM HEPES pH7.4, 50 mM NaCl, 5% Glycerol, 1 mM DTT and 1 mM CHAPS. Reference Compound X was diluted, in assay buffer containing 20 nM single mutant, tandem bromodomain protein, to a concentration equivalent to 2*Kd for this bromodomain. The solution containing bromodomain and Reference Compound X was added to dose response dilutions of test compound or DMSO vehicle (a maximum of 0.5% DMSO is used in this assay) in Greiner 384 well black low volume microtitre plates and subsequently incubated for 30 minutes at rt. An equal volume of 3 nM of anti-6*His Europium chelate was added to all wells, followed by a further 30 minute incubation at rt. TR-FRET was detected using a Perkin Elmer Multimode plate reader, by exciting the donor fluorophore at λ337 nm and subsequently, after a delay of 50 μsecs, measuring emission of the donor and acceptor fluorophores at λ615 nm and λ665 nm, respectively. In order to control these assays, 16 replicates each of uninhibited (DMSO vehicle) and inhibited (10*IC50 concentrations of Example 11 of WO 2011/054846A1) TR-FRET assays were included on every microtitre plate.
cA four parameter curve fit of the following form was then applied:
y=a+((b−a)/(1+(10{circumflex over ( )}x/10{circumflex over ( )}c){circumflex over ( )}d)
Where ‘a’ is the minimum, ‘H’ is the Hill slope, ‘c’ is the pIC50 and ‘d’ is the maximum.
All compounds (Examples) were each tested in the BRD4 BD1 and the BRD4 BD2 TR-FRET assays essentially as described above. Those of skill in the art will recognise that in vitro binding assays and cell-based assays for functional activity are subject to experimental variability. Accordingly, it is to be understood that the pIC50 values given below are exemplary only. pIC50 values are expressed as log10 units.
All Examples, with the exception of Examples 131, 242-247 and 249-261, were found to have a pIC50≥5.0 in at least one assay described above.
Examples 94, 96, 102, 106, 108-110, 112, 114, 132, 135, 135, 136, 137, 229, 233, 234, 236-241 and 248. were found to have a pIC50≥ 5.0 and <6.0 in the BRD4 BD2 assay.
All other compounds were found to have a pIC50≥6.0 and <8.0 in the BRD4 BD2 assay. In particular, Example 1 was found to have a pIC50 of 7.2 in the BRD4 BD2 assay; Example 2 was found to have a pIC50 of 7.1 in the BRD4 BD2 assay; Example 3 was found to have a pIC50 of 7.5 in the BRD4 BD2 assay; and Example 139 was found to have a pIC50 of 7.4 in the BRD4 BD2 assay.
Calculation of Selectivity for BRD4 BD2 Over BRD4 BD1
Selectivity for BRD4 BD2 over BRD4 BD1 was calculated as follows:
Selectivity=BRD4 BD2 pIC50−BRD4 BD1 pIC50 pIC50
All Examples, with the exemption of Examples 114, 116, 131, 134, 136-138 and 238-261, were found to have selectivity for BRD4 BD2 over BRD4 BD1 of ≥1 log unit in at least one of the TR-FRET assays described above, and hence are at least 10 fold selective for BRD4 BD2 over BRD4 BD1.
Examples 1-91, 139-219 were found to have selectivity for BRD4 BD2 over BRD4 BD1 of ≥2 log unit in at least one of the TR-FRET assays described above, and hence are at least 100 fold selective for BRD4 BD2 over BRD4 BD1.
Example 1 was found to have a selectivity for BRD4 BD2 over BRD4 BD1 of 2.8 log units in at least one of the TR-FRET assays described above.
Example 2 was found to have a selectivity for BRD4 BD2 over BRD4 BD1 of 3.0 log units in at least one of the TR-FRET assays described above.
Example 3 was found to have a selectivity for BRD4 BD2 over BRD4 BD1 of 3.0 log units in at least one of the TR-FRET assays described above.
Example 139 was found to have a selectivity for BRD4 BD2 over BRD4 BD1 of 3.0 log units in at least one of the TR-FRET assays described above.

Claims (17)

The invention claimed is:
1. A compound of formula (I)
Figure US10966961-20210406-C00483
or a pharmaceutically acceptable salt thereof
wherein:
R1 is —C1-3alkyl or cyclopropyl;
R2 is —C0-3alkyl-cycloalkyl, wherein the cycloalkyl group is optionally substituted with one, two, or three R5 groups which may be the same or different;
or R2 is —C0-4alkyl-heterocyclyl or —(CH2)pO-heterocyclyl, wherein each heterocyclyl is optionally substituted by one or two R9 groups which may be the same or different; or
R2 is H, —CH3, —C2-6alkyl optionally substituted by one, two, three, four or five fluoro, —C2-6alkylOR6, —C2-6alkylNR10aR11a, —(CH2)mSO2C1-3alkyl, —(CH2)mSO2NR10R11, (CH2)mC(O)NR10R11, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mNHCO2C1-4alkyl, —(CH2)mNHC(O)C1-4alkyl, or —(CH2)nheteroaryl, wherein the heteroaryl is optionally substituted by one or two R7 groups which may be the same or different;
R3 is H, —C1-4alkyl, cyclopropyl, —CH2F, —C1-3alkylOR6, or —C1-3alkylCN;
R4 is phenyl or a heteroaryl group, wherein each are optionally substituted by one, two, or three R7 groups which may be the same or different;
each R5 is independently halo, —C0-6alkyl-R8, —OCH2phenyl, —CN, or —SO2C1-3alkyl;
R6 is H or —C1-4alkyl;
each R7 is independently oxo, halo, —C1-4alkyl optionally substituted by one, two or three fluoro, —C0-3alkylOR6, —OC2-3alkylOR6, —C0-3alkylNR10R11, —C0-3alkyl-CONR10R11, —SO2—C1-3alkyl,—SO2NR10R11, or —SO2phenyl optionally substituted by —C1-4alkyl;
R8 is H, —OR6, —NR10R11 heteroaryl;
each R9 is independently halo, —C1-4alkyl, cyclopropyl, cyclobutyl, —CH2CF3, —CH2CHF2, —CH2CH2F, —OCH2CH2OR6, —C0-3alkylOR6, —C0-3alkylNR10R11, —NHCH2CH2OR6, —NHCO2C1-4alkyl, oxo, —C(O)R6, —C(O)OR6, or —C(O)NR10R11,
R10 and R11 are each independently selected from H and —C1-3alkyl; or R10 and R11 may join together with the nitrogen to which they are attached, to form a 4 to 7-membered heterocyclyl optionally substituted by one or two substituents independently selected from —C1-3alkyl optionally substituted with one, two, or three fluorine atoms, —C2-4alkylOH, —OH, and F;
R10a and R11a are each independently selected from H and —C1-3alkyl;
m is an integer selected from 2, 3, or 4;
n is an integer selected from 0, 1, 2, 3, or 4; and
p is an integer selected from 2, 3, or 4.
2. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R1 is methyl.
3. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R2 is a —C0-3alkyl-C3-7cycloalkyl group, wherein the C3-7cycloalkyl group is selected from cyclopropyl, cyclobutyl, cyclohexyl, and bicyclo[3.1.0]hexanyl, wherein said cyclopropyl, cyclobutyl, cyclohexyl, or bicyclo[3.1.0]hexanyl is optionally substituted with one, two, or three R5 groups which may be the same or different.
4. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R2 is —C0-4alkyl-heterocyclyl wherein the heterocyclyl is selected from oxetanyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, morpholinyl, piperidinyl, piperazinyl, (1r,5s)-3-oxabicyclo[3.1.0]hexanyl and (1r,5s)-3-azabicyclo[3.1.0]hexanyl, wherein said oxetanyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, morpholinyl, piperidinyl, piperazinyl, (1r,5s)-3-oxabicyclo[3.1.0]hexanyl or (1r,5s)-3-azabicyclo[3.1.0]hexanyl is optionally substituted by one or two R9 groups which may be the same or different.
5. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R2 is methyl, ethyl, propyl, iso-propyl, butyl, —CH2CH2CH(CH3)2, —CH2CH(CH3)2, —CH2CH2OR6, —CH2CH2CH2OR6, —CH2CH(CH3)OR6, —CH2CH2CH(CH3)OR6, —CH2CH2CH(CH3)NR10R11, —CH2CH2CH2NR10R11, —(CH2)mSO2CH3, —(CH2)mC(O)NHCH3, —(CH2)mCN, —(CH2)mCO2R6, —(CH2)mCF3, and —(CH2)mNHCO2C(CH3)3.
6. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R2 is —(CH2)nC5-6heteroaryl, wherein the C5-6heteroaryl group is selected from furanyl, thienyl, pyrrolyl, triazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridinyl, pyridazinyl, pyrazinyl, and pyrimidinyl, wherein said furanyl, thienyl, pyrrolyl, triazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridinyl, pyridazinyl, pyrazinyl or pyrimidinyl is optionally substituted by one or two substituents independently selected from halo, C1-4alkyl, C3-4cycloalkyl, and —C0-3alkylOR6.
7. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R3 is H, methyl, ethyl, —CH2F, —CH2OH, —CH(OH)CH3, —CH2OMe, or —CH2CN.
8. The compound or pharmaceutically acceptable salt thereof according to claim 1, wherein R4 is unsubstituted phenyl or is phenyl substituted by one or two R7 groups which may be the same or different selected from halo, —C1-4alkyl, —C0-3alkylOR6, and CN.
9. The compound or salt thereof according to claim 1, wherein R4 is a heteroaryl group selected from the group consisting of pyridyl, indolyl, and pyrrolopyridinyl, wherein said pyridyl, indolyl, and pyrrolopyridinyl is optionally substituted by one, two, or three R7 groups which may be the same or different.
10. A compound which is selected from
N5-((1r,4S)-4-hydroxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide;
N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-1-((S)-1-(4-chlorophenyl)ethyl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide;
(S)—N3-methyl-1-(1-phenylethyl)-N5-(1H-pyrazol-4-yl)-1H-pyrazole-3,5-dicarboxamide;
1-((S)-1-(4-chlorophenyl)ethyl)-N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide;
1-((S)-1-(3-chlorophenyl)ethyl)-N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1H-pyrazole-3,5-dicarboxamide;
N5-((1R,3R,5S,6r)-3-hydroxybicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylpropyl)-1H-pyrazole-3,5-dicarboxamide; and
N5-((1r,4S)-4-methoxycyclohexyl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide
or a pharmaceutically acceptable salt thereof.
11. A compound which is N5-((1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl)-N3-methyl-1-((S)-1-phenylethyl)-1H-pyrazole-3,5-dicarboxamide represented by the formula
Figure US10966961-20210406-C00484
or a pharmaceutically acceptable salt thereof.
12. A pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof according to claim 1 and one or more pharmaceutically acceptable excipients.
13. A combination comprising the compound or pharmaceutically acceptable salt thereof according to claim 1 together with one or more other therapeutically active agents.
14. A method of treatment of a disease or condition in a human for which a bromodomain inhibitor is indicated, the method comprising administering to the human in need thereof a therapeutically effective amount of the compound or pharmaceutically acceptable salt thereof according to claim 1, wherein the disease or condition is an acute or chronic autoimmune and/or inflammatory condition.
15. A method of treatment according to claim 14, wherein the disease or condition involves an inflammatory response to an infection with bacteria, a virus, fungi, a parasite, or their toxins.
16. A method of treatment according to claim 14, wherein the disease or condition is a viral infection.
17. A method of treatment according to claim 14, wherein the disease or condition is rheumatoid arthritis.
US16/489,859 2017-03-01 2018-02-27 Pyrazole derivatives as bromodomain inhibitors Active US10966961B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB1716374 1974-04-19
GB1703283 2017-03-01
GBGB1703283.0A GB201703283D0 (en) 2017-03-01 2017-03-01 Compounds
GB1703283.0 2017-03-01
GB1716374.2 2017-10-06
GBGB1716374.2A GB201716374D0 (en) 2017-10-06 2017-10-06 Compounds
PCT/EP2018/054733 WO2018158212A1 (en) 2017-03-01 2018-02-27 Pyrazole derivatives as bromodomain inhibitors

Publications (2)

Publication Number Publication Date
US20190381010A1 US20190381010A1 (en) 2019-12-19
US10966961B2 true US10966961B2 (en) 2021-04-06

Family

ID=61691917

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/489,859 Active US10966961B2 (en) 2017-03-01 2018-02-27 Pyrazole derivatives as bromodomain inhibitors

Country Status (22)

Country Link
US (1) US10966961B2 (en)
EP (1) EP3589618B1 (en)
JP (1) JP6857254B2 (en)
KR (1) KR102403176B1 (en)
CN (1) CN110582485B (en)
AU (1) AU2018228651B2 (en)
BR (1) BR112019018028A2 (en)
CA (1) CA3054754A1 (en)
CL (1) CL2019002480A1 (en)
CO (1) CO2019009020A2 (en)
CR (1) CR20190391A (en)
DO (1) DOP2019000221A (en)
ES (1) ES2938269T3 (en)
IL (1) IL268565B (en)
JO (1) JOP20190192A1 (en)
MX (1) MX2019010218A (en)
PE (1) PE20191535A1 (en)
PH (1) PH12019501999A1 (en)
SG (1) SG11201907546WA (en)
TW (1) TW201841893A (en)
UY (1) UY37622A (en)
WO (1) WO2018158212A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201814167D0 (en) * 2018-08-31 2018-10-17 Glaxosmithkline Ip No 2 Ltd Compounds
CN113402428B (en) * 2021-06-11 2023-03-03 重庆医药高等专科学校 Preparation method of trans-4- (tert-butoxycarbonylamino) cyclohexanecarboxylic acid and intermediate thereof
WO2024018423A1 (en) 2022-07-21 2024-01-25 Tay Therapeutics Limited Pyrroles and imidazoles as bet protein inhibitors

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1357111A1 (en) 2000-12-28 2003-10-29 Shionogi & Co., Ltd. Pyridone derivative having affinity for cannabinoid 2-type receptor
WO2004033446A1 (en) 2002-10-09 2004-04-22 Danter Wayne R Protein tyrosine kinase inhibitors
EP1433788A1 (en) 2002-12-23 2004-06-30 Aventis Pharma Deutschland GmbH Pyrazole-derivatives as factor Xa inhibitors
EP1477186A1 (en) 2002-02-19 2004-11-17 Shionogi & Co., Ltd. Antipruritics
US7910606B2 (en) * 2002-12-23 2011-03-22 Sanofi-Aventis Deutschland Gmbh Pyrazole-derivatives as factor Xa inhibitors
US20120208814A1 (en) 2009-11-05 2012-08-16 Emmanuel Hubert Demont Thetrahydroquinolines Derivatives As Bromodomain Inhibitors
WO2014074675A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company Heteroaryl substituted pyridyl compounds useful as kinase modulators
WO2014096965A2 (en) 2012-12-21 2014-06-26 Rvx Therapeutics Inc. Novel heterocyclic compounds as bromodomain inhibitors
WO2015015318A2 (en) 2013-07-31 2015-02-05 Zenith Epigenetics Corp. Novel quinazolinones as bromodomain inhibitors
WO2017037116A1 (en) 2015-09-02 2017-03-09 Glaxosmithkline Intellectual Property (No.2) Limited Pyridinone dicarboxamide for use as bromodomain inhibitors
WO2017060180A1 (en) 2015-10-05 2017-04-13 Glaxosmithkline Intellectual Property (No.2) Limited 2-oxo-1,2-dihydropyridine-3,5-dicarboxamide compounds as bromodomain inhibitors
WO2017174621A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Pyridyl derivatives as bromodomain inhibitors
WO2017202742A1 (en) 2016-05-24 2017-11-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridine dicarboxamide derivatives as bromodomain inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE60329529D1 (en) * 2002-12-23 2009-11-12 Sanofi Aventis Deutschland Pyrazoline Derivatives as Factor Xa Inhibitors
CN101490273A (en) * 2006-05-09 2009-07-22 南卡罗来纳州医科大学研究发展基金会 Detecting diastolic heart failure by protease and protease inhibitor plasma profiling
DK2401270T3 (en) * 2009-02-26 2014-01-13 Glaxo Group Ltd Pyrazole derivatives used as CCR4 receptor antagonists.
AR084070A1 (en) * 2010-12-02 2013-04-17 Constellation Pharmaceuticals Inc BROMODOMINIUM INHIBITORS AND USES OF THE SAME
CN105523955B (en) * 2015-12-14 2018-08-17 北京嘉林药业股份有限公司 Compound and its purposes in medicine preparation

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1357111A1 (en) 2000-12-28 2003-10-29 Shionogi & Co., Ltd. Pyridone derivative having affinity for cannabinoid 2-type receptor
EP1477186A1 (en) 2002-02-19 2004-11-17 Shionogi & Co., Ltd. Antipruritics
WO2004033446A1 (en) 2002-10-09 2004-04-22 Danter Wayne R Protein tyrosine kinase inhibitors
EP1433788A1 (en) 2002-12-23 2004-06-30 Aventis Pharma Deutschland GmbH Pyrazole-derivatives as factor Xa inhibitors
US7910606B2 (en) * 2002-12-23 2011-03-22 Sanofi-Aventis Deutschland Gmbh Pyrazole-derivatives as factor Xa inhibitors
US20120208814A1 (en) 2009-11-05 2012-08-16 Emmanuel Hubert Demont Thetrahydroquinolines Derivatives As Bromodomain Inhibitors
WO2014074675A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company Heteroaryl substituted pyridyl compounds useful as kinase modulators
WO2014096965A2 (en) 2012-12-21 2014-06-26 Rvx Therapeutics Inc. Novel heterocyclic compounds as bromodomain inhibitors
US20140179648A1 (en) 2012-12-21 2014-06-26 Rvx Therapeutics Inc. Novel Heterocyclic Compounds as Bromodomain Inhibitors
WO2015015318A2 (en) 2013-07-31 2015-02-05 Zenith Epigenetics Corp. Novel quinazolinones as bromodomain inhibitors
WO2017037116A1 (en) 2015-09-02 2017-03-09 Glaxosmithkline Intellectual Property (No.2) Limited Pyridinone dicarboxamide for use as bromodomain inhibitors
WO2017060180A1 (en) 2015-10-05 2017-04-13 Glaxosmithkline Intellectual Property (No.2) Limited 2-oxo-1,2-dihydropyridine-3,5-dicarboxamide compounds as bromodomain inhibitors
WO2017174621A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Pyridyl derivatives as bromodomain inhibitors
WO2017202742A1 (en) 2016-05-24 2017-11-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridine dicarboxamide derivatives as bromodomain inhibitors

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
Dittmer et al., "Models for the Pyridine Nucleotide Coenzymes. Synthesis and Properties of Bridged Dinicotinamide Derivatives1-3", J. Org. Chem., vol. 38, No. 16, pp. 2873-2882 (1973).
Gallenkamp et al., "Bromodomains and Their Pharmacological Inhibitors", ChemMedChem, vol. 9, No. 3, pp. 438-464 (2014).
Gamier et al., "BET bromodomain inhibitors: a patent review", Expert Opinion on Therapeutic Patents, vol. 24, No. 2, pp. 185-199 (2014).
International Search Report for International application No. PCT/EP2016/070519, dated Oct. 20, 2016, 4 pages.
International Search Report for International application No. PCT/EP2016/072216, International filing date: Sep. 20, 2016, 3 pages.
International Search Report for International application No. PCT/EP2016/073532, dated Nov. 30, 2016, 5 pages.
International Search Report for International application No. PCT/EP2017/058050, dated May 24, 2017, 5 pages.
International Search Report for International application No. PCT/EP2017/062208, dated Jul. 6, 2017, 5 pages.
International Search Report for International application No. PCT/EP2018/054730, dated May 4, 2018, 5 pages.
International Search Report for International application No. PCT/EP2018/054733, dated Jun. 11, 2018, 5 pages.
Non-Final Office Action for U.S. Appl. No. 15/766,222, USPTO, notification dated Oct. 4, 2018, 9 pages.
Notice of Allowance for U.S. Appl. No. 15/762,229, USPTO, dated Dec. 11, 2018, 9 pages.
Notice of Allowance for U.S. Appl. No. 15/762,229, USPTO, dated Mar. 20, 2019, 9 pages.
Notice of Allowance for U.S. Appl. No. 15/766,222, USPTO, dated Jan. 17, 2019, 6 pages.
Restriction Requirement for U.S. Appl. No. 15/757,199, USPTO, notification dated Feb. 11, 2019, 9 pages.

Also Published As

Publication number Publication date
EP3589618A1 (en) 2020-01-08
IL268565A (en) 2019-09-26
CA3054754A1 (en) 2018-09-07
CO2019009020A2 (en) 2019-08-30
JP6857254B2 (en) 2021-04-14
CR20190391A (en) 2019-10-17
IL268565B (en) 2022-06-01
US20190381010A1 (en) 2019-12-19
PE20191535A1 (en) 2019-10-23
MX2019010218A (en) 2019-10-21
KR102403176B1 (en) 2022-05-27
SG11201907546WA (en) 2019-09-27
JOP20190192A1 (en) 2019-08-08
CL2019002480A1 (en) 2019-11-29
UY37622A (en) 2018-09-28
CN110582485B (en) 2023-10-13
EP3589618B1 (en) 2022-12-28
CN110582485A (en) 2019-12-17
JP2020509033A (en) 2020-03-26
WO2018158212A1 (en) 2018-09-07
AU2018228651B2 (en) 2020-12-17
AU2018228651A1 (en) 2019-08-29
TW201841893A (en) 2018-12-01
KR20190123307A (en) 2019-10-31
PH12019501999A1 (en) 2020-06-15
DOP2019000221A (en) 2019-11-15
ES2938269T3 (en) 2023-04-05
BR112019018028A2 (en) 2020-03-24

Similar Documents

Publication Publication Date Title
US10370356B2 (en) Pyridinone dicarboxamide for use as bromodomain inhibitors
US10844015B2 (en) Pyridine dicarboxamide derivatives as bromodomain inhibitors
US10927080B2 (en) Pyridinone dicarboxamide for use as bromodomain inhibitors
US10934272B2 (en) Pyridyl derivatives as bromodomain inhibitors
US10471050B2 (en) 2-OXO-1,2-dihydropyridine-3,5-dicarboxamide compounds as bromodomain inhibitors
US10583112B2 (en) Benzo[b]furans as bromodomain inhibitors
US10966961B2 (en) Pyrazole derivatives as bromodomain inhibitors
US11273146B2 (en) Benzofuran derivatives and their use as bromodomain inhibitors
US20210346336A1 (en) Furan derivatives as bromodomain inhibitors
US10849897B2 (en) Pyridyl derivatives as bromodomain inhibitors
US11319307B2 (en) 2,3-dihydrobenzohurans as bromodomain inhibitors
EA038888B1 (en) Pyrazole derivatives as bromodomain inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMIT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ATKINSON, STEPHEN JOHN;DEMONT, EMMANUEL HUBERT;HARRISON, LEE ANDREW;AND OTHERS;REEL/FRAME:050212/0668

Effective date: 20180312

Owner name: GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ATKINSON, STEPHEN JOHN;DEMONT, EMMANUEL HUBERT;HARRISON, LEE ANDREW;AND OTHERS;REEL/FRAME:050212/0668

Effective date: 20180312

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE